You are on page 1of 593

CHURCHILL LIVINGSTONE

An Imprint of Elsevier
The Curtis Center
Independence Square West
Philadelphia, Pennsylvania 19106

Copyright © 2004, 1980, Elsevier Inc. (USA). All Rights Reserved

No part of this publication may be reproduced or transmitted in any form or by any means,
electronic or mechanical, including photocopy, recording, or any information storage and
retrieval system, without permission in writing from the publisher.

Permissions may be sought directly from Elsevier’s Health Sciences Rights Department
in Philadelphia, USA: phone: (+1)215 238 7869, fax: (+1)215 238 2239, email:
healthpermissions@elsevier.com. You may also complete your request on-line via the Elsevier Science
homepage (http://www.elsevier.com, by selecting “Customer Support” and then
“Obtaining Permissions.”

CHURCHILL LIVINGSTONE and the the Sail Boat Design are trademarks of Harcourt, Inc., registered in the
United States of America and/or other jurisdictions.

Notice
Medicine is an ever-changing field. Standard safety precautions must be followed, but as new
research and clinical experience broaden our knowledge, changes in treatment and drug therapy
may become necessary or appropriate. Readers are advised to check the most current product
information provided by the manufacturer of each drug to be administered to verify the
recommended dose, the method and duration of administration, and contraindications. It is the
responsibility of the treating physician, relying on experience and knowledge of the patient, to
determine dosages and the best treatment for each individual patient. Neither the Publisher nor
the editor assumes any liability for any injury and/or damage to persons or property arising from
this publication.
The Publisher

Library of Congress Cataloging-in-Publication Data


Petz, Lawrence D.
Immune hemolytic anemias / Lawrence D. Petz, George Garratty.—2nd ed.
p. ; cm.
Rev. ed. of: Acquired immune hemolytic anemias. 1980.
Includes bibliographical references.
ISBN 0-443-08559-5
1. Hemolytic anemia, Autoimmune. I. Garratty, George. II. Petz, Lawrence D. Acquired
immune hemolytic anemias. III. Title.
[DNLM: 1. Anemia, Hemolytic, Autoimmune. WH 170 P513i 2004]
RC641.7.H4P47 2004
616.1′52—dc21
2003043767

Printed in United States of America

Last digit is the print number: 9 8 7 6 5 4 3 2 1


Preface to the Second Edition
We thought we might have set a record for the longest time between editions of a
book, since the first edition of this book, entitled Acquired Immune Hemolytic Anemias,
was published 24 years ago. However, our mentor, Professor Sir John Dacie, published
the third edition of Autoimmune Haemolytic Anaemias (Volume 3 of The Haemolytic
Anemias) in 1992, just 30 years after the publication of the previous edition of that
volume!
We have been flattered during these long years by a number of physicians,
immunohematologists, and blood bankers who insist that they still use the first edition
and have continued to press us for the second. As with the first edition, this book is
intended primarily as a useful source of information for those who care for patients
who have immune hemolytic anemias, that is, clinicians with patient care responsibil-
ity and blood bank professionals, including physicians and technical staff. However,
this purpose cannot be properly served without an adequately detailed scientific back-
ground, and we have endeavored to supply this. We have attempted to be rather com-
prehensive, but we do not intend this book to be only a reference volume and have
therefore included practical aspects of the evaluation and management of patients with
hemolysis.
Patients with immune hemolytic anemias are sufficiently common as to constitute
an important problem, but, on the other hand, they are sufficiently unusual that it is dif-
ficult for many individuals outside of referral centers to acquire adequate experience to
feel at ease in managing the multitude of problems such patients may present. We
earnestly hope that sharing our experiences through the medium of this book will be of
value to others who confront such problems less commonly.
During the years between editions of this text, medical disciplines that were rather
early in their developmental stages, such as hematopoietic cell and solid organ trans-
plantation, have emerged to be major components of health care and have contributed
to the emergence of entirely new causes of immune hemolysis. Also, new “generations”
of drugs have been developed, one of the consequences of which is an expansion of the
causes of drug-induced immune hemolytic anemias. Molecular biology and DNA
technology have evolved to become a part of our everyday scientific lives and are being
utilized in hematology as in all other disciplines. We have attempted to bring our first
edition up to date while not ignoring important earlier contributions. We have added
chapters on Historical Concepts of Immune Hemolytic Anemias, Hemolytic Disease of
the Fetus and Newborn, Immune Hemolysis Associated with Transplantation, and
Hemolytic Transfusion Reactions.
As we emphasized in the preface to the first edition, one of the important aspects
of diagnosis and management of patients with immune hemolytic anemias is that the
care of such patients depends on a knowledge of some aspects of both clinical and
laboratory medicine. Although this is true throughout clinical medicine, a problem of
particular magnitude is created by the need for clinicians to be able to interpret such
unusual laboratory tests as the direct antiglobulin test with monospecific antiglobulin
sera and the specificity and thermal range of allo- and autoantibodies. Similarly,
vi Preface to the Second Edition

laboratory personnel should be able to assist clinicians in the interpretation of import-


ant data, as when transfusion is indicated for a patient whose serum reacts with all
RBCs in compatibility tests. Accordingly, one of the primary purposes of this book is to
present both the clinical and laboratory aspects of immune hemolytic anemias in a
single volume. We strongly feel that neither laboratory personnel (including physi-
cians) nor clinicians can optimally contribute to the care of patients with immune
hemolytic anemias without a firm understanding of both aspects of the subject.

Lawrence D. Petz
George Garratty
Preface to the First Edition
This book is intended to be a useful source of information for those who care for
patients who have immune hemolytic anemias, i.e., clinicians with primary responsi-
bility for patient management, physicians concerned with laboratory medicine, includ-
ing blood bank directors, and the technical staff of such laboratories. It is not intended
as an encyclopedic review or as a “tour de force” exposition of facts that are of interest
primarily to those with extensive background and a highly specialized interest in the
field.
Patients with immune hemolytic anemias are sufficiently common as to constitute
an important problem but, on the other hand, are sufficiently unusual that it is difficult
for many individuals outside of referral centers to acquire adequate experience to feel
at ease in managing the multitude of problems such patients may present. We have had
a special interest in these disorders and we earnestly hope that sharing our experiences
through the medium of this book will be of value to others who confront such problems
less commonly. We include previously unpublished data concerning our experiences
with various phases of the diagnosis and management of more than 300 patients, as
well as a review of relevant information available in the medical literature.
Although the primary purpose of this book is, therefore, to be a source of informa-
tion that will be of value in management of patients, this purpose cannot be adequately
served merely by a superficial exposition of “practical” facts, and we do not intend this
book to be a manual of patient care. We trust that the interested reader would demand
an adequately detailed scientific background to make meaningful the recommended
laboratory procedures and their clinical interpretation. For example, the knowledge
that the direct antiglobulin (Coombs’) test performed on red cells from patients with
cold agglutinin syndrome is invariably positive using anit-C3d antiglobulin serum and
invariably negative using anti-IgG antiglobulin serum is of some clinical value (Ch. 6).
When such information is augmented by an understanding of pertinent aspects of the
serum complement system (Ch. 3) and the mechanisms of immune hemolysis (Ch. 4),
one then has a basis for understanding such facts and their clinical significance.
Writing this book presents a unique problem. That is, one of the important aspects
of diagnosis and management of patients with immune hemolytic anemias is that the
care of such patients depends upon a knowledge of some aspects of both clinical and
laboratory medicine. Although this is true throughout medicine, a problem of unusual
magnitude is created by the fact that most clinicians have very little exposure to
immunohematology. Results of direct antiglobulin tests with monospecific antiglobulin
sera and the characterization of serum antibody specificity and thermal range is
information that is difficult or impossible for most practicing physicians to utilize. This
problem is augmented by the fact that laboratory personnel are faced with difficult
technical tasks, and, in the very best of hands, uncertainties may remain. For example,
in regard to blood transfusion (Ch. 10), what is the probability of not detecting a red cell
alloantibody in the serum of a patient with autoimmune hemolytic anemia when the
serum reacts with all donor cells tested, and what is the risk of transfusion of blood that
is incompatible because of the presence of an autoantibody? One of the prime purposes
viii Preface to the First Edition

of this book, and one of the more difficult tasks we faced in writing it, is to present both
the laboratory and clinical aspects of immune hemolytic anemias in a single volume in
a manner that is understandable by those in both fields. Neither laboratory personnel
(including physicians) nor clinicians can optimally contribute to the care of patients
with immune hemolytic anemias without an understanding of both aspects of the
subject. Therefore, it is our firm opinion that, with few exceptions (e.g., some sections
concerning technical details which may justifiably be ignored by clinicians, and some
aspects of therapy which may not be essential knowledge for technologists), the infor-
mation herein is important to those in both clinical and laboratory medicine for proper
management of patients with immune hemolytic anemias.

Lawrence D. Petz
George Garratty
Acknowledgments
As indicated in the first edition, we are both indebted to Professor Sir John Dacie
for the privilege of working in his laboratory at the Royal Postgraduate Medical School
and Hammersmith Hospital in London. His teachings served as a foundation for our
work and, moreover, we have attempted to emulate his dedication and precision in
scientific investigation.
Grateful acknowledgment is also due to the numerous physicians and technolo-
gists who were kind enough to refer interesting and challenging clinical and laboratory
problems to us. Without this continued support it would have been impossible to
acquire the experience and data necessary to compile this volume. In addition, we
appreciate the collaboration of our colleagues at the City of Hope Medical Center,
Duarte, California, and the University of California Los Angeles Medical Center
(Dr. Petz) and American Red Cross Blood Services, Southern California Region
(Dr. Garratty).
We would especially like to thank some extraordinary medical technologists who
were not just a “pair of hands” in the laboratory but were innovative contributors to the
design and results of our studies: Donald Branch (now the proud possessor of a PhD);
Alana (Loni) Calhoun, Patricia Arndt, Regina Leger, Sandra Nance, and Nina Postoway.
Their relevant roles were obvious from our publications mentioned throughout the
book. Dr. Garratty would especially like to thank Ann Tunick, his administrative assist-
ant (since 1978), who typed multiple error-free drafts of material, found and formatted
references, and dealt imperturbably with all problems that arose. Without her help
Dr. Garratty’s contributions would never have appeared in this book!
Both of us would like to acknowledge the tremendous support of our wives
(Thelma Petz and Eileen Garratty), who put up with our working every weekend and
many evenings without too many grumbles!
C H A P T E R 1

Historical Concepts
of Immune Hemolytic
Anemias

Immune hemolysis is a short- process of agglutination, understanding the distinction


ening of red blood cell (RBC) between congenital and acquired disorders, under-
survival due, directly or indi- standing that premature destruction of RBCs can cause
rectly, to antibodies. These anti- anemia and jaundice, recognizing spherocytes and
bodies may be autoantibodies abnormal osmotic fragility of RBCs and determining
or alloantibodies. This chapter their significance in patients with hemolysis, recogniz-
will deal mainly with historical ing reticulocytes, determining that serum antibodies
aspects of autoimmune hemo- may cause destruction of foreign cells and also autolo-
lytic anemia (AIHA), followed gous cells, developing means to measure RBC survival,
by a brief discussion of histori- developing diagnostic assays for antibodies, refuting
cal aspects of hemolytic transfusion reactions. the concept of horror autotoxicus, and understanding the
AIHA is an acquired immunologic disease in which role of the spleen and splenectomy.
the patient’s RBCs are selectively attacked and The discoveries that led to the development of our
destroyed (hemolysed) by autoantibodies produced knowledge about these concepts are herein reviewed
by the patient’s own immune system. Shortened RBC in the approximate order in which the relevant obser-
survival is frequently associated with the presence of vations were made. Here, then, is how our knowledge
a reticulocytosis, spherocytes in the peripheral blood of AIHA came to be. The development of this short
film, autoantibodies in the patient’s serum, and occa- review was aided significantly by previous reviews on
sionally splenomegaly, hemoglobinemia, and hemo- various aspects of hemolysis and AIHA.1-9
globinuria. Although these facts are common
knowledge now, it was not always so. Reviewing how
these concepts developed over the centuries by obser- THE LESSONS OF HISTORY
vation and clinical and laboratory experimentation is
both fascinating and instructive. Everyone who studies the stories of discovery in what
It is evident that concepts that collectively led to our has come to be called the field of hematology will rec-
present understanding of AIHA required knowledge of ognize the early gropings in the midst of profound
the existence of RBCs, understanding the possibility of ignorance and the difficulties that confronted the
anemia without blood loss, distinguishing hemoglo- investigators. We have gained an understanding of
binuria from hematuria, understanding the mechanism biology that could hardly have been dreamed of only
by which hemoglobinuria occurs, recognizing the a short time ago, let alone at the time of the first
1
2 Immune Hemolytic Anemias

tentative forays into the unknown. Moreover, under- did they fail to make mistakes. Indeed, incorrect
standing has been crowned by tangible benefits for theories have hampered the advance of knowledge,
humanity. It is worthwhile to consider how such great especially when these theories were widely dissemin-
progress comes about and why. How is knowledge ated and were pronounced by eminent authorities. A
achieved, and what can we learn from the process by number of such examples appear in the following
which important discoveries were made?10 pages.
The first lesson to be learned of history is that the It follows that authorities must be humble and
path of progress is anything but straight. The course novices skeptical.
of research has been likened to the flow of a stream
that ultimately becomes a rushing torrent whose
importance is obvious. This certainly has been the
history of research in hematology.
EARLIEST DESCRIPTIONS OF POSSIBLE
It certainly does not follow that, because a concept ACQUIRED HEMOLYTIC ANEMIA
is plausible and is in accord with the understanding of
the time, it is necessarily correct. The following pages The first written description of what may have been
provide many examples of misinterpretations result- an acquired hemolytic anemia, albeit not of an
ing from such an assumption. Furthermore, because immune nature, was Galen’s description in 150 AD of
they have been plausible, such views often have a person bitten by a viper whose “skin turned the
endured and have stood in the way of acceptance of color of a ripe leek.”1,4,11 Galen’s understanding of
observations and interpretations that proved to be the physiology was such that he implicated the spleen as
correct ones. leading to the skin discoloration, an association of the
Discovery begins with an observation or the posing spleen and hemolysis that was not confirmed until the
of a question. But observation is not as simple as it late nineteenth century.1
sounds. Indeed, many look but few see. It is the excep- PCH may have been described as early as 1529 by
tional person who recognizes the unusual event or Johannes Actuarius, a court physician in Constan-
manifestation. Still fewer pursue it to new under- tinople. In his work, De Urinis, Acturarius described a
standing. Many may ask questions but few have the condition in which the urine is “azure & livid as well
imagination, the energy, and the overpowering drive as black” in patients being of melancholic humor and
to persist in the search for an answer, especially when complaining of loss of strength, after an exposure to
this must be done in the face of difficulties and fail- cold.4 Further mention of PCH seems, however, to be
ures and even despite scorn from their peers. absent for nearly 300 years, until the latter half of the
Imagination and industry alone, however, have not nineteenth century.
sufficed. Means have had to be devised to explore the
questions that were posed. When these were provided,
it is impressive to see what the introduction of a new EARLY EXPERIMENTAL
technique made possible for an area of inquiry. A
simple example, described later, is the introduction of INVESTIGATION OF BLOOD
the antiglobulin test, which very rapidly led to a much
clearer distinction between immune and nonimmune Description of Red Blood Cells. The development of
hemolytic anemias. the scientific method led to the seminal discoveries of
Progress depends on the contributions of many. the circulation of blood by Harvey in the early sixteenth
Moreover, scientific discipline has benefited from century and the cardinal experiments with transfusion
developments in other fields, progress in one field of blood by Lower in England and Denis in Paris in the
spurring another, and vice versa. As knowledge has mid-seventeenth century. Despite this interest in blood,
grown, it has become impossible for a single human the discovery of the RBCs had to await the appearance
being to encompass the whole, and the discovery and of the microscope around 1650. The first observation of
growth of understanding have become more and more an RBC was likely made by Malpighi in 1661, when he
dependent on interchange among scientific disciplines. described the circulation of RBCs in the capillaries, and
Still another aspect of the progress of understand- this was followed in 1663 by Swammerdan’s descrip-
ing is worth noting. It is not generally appreciated tion of minute globules in the blood of a frog. A decade
how often curiosity concerning an observation made later, human RBCs were described in detail by van
at the bedside by clinicians has led to far-reaching Leeuwenhoek (Fig. 1-1),12 who also established their
investigations. An example is the observation size at about 1⁄3000 of an inch by comparing an RBC with
of hemoglobinuria, which led to the understanding of a grain of sand of known size.
destruction of RBCs and to the early delineation of John Huxham, in 1770, described the changing
certain clinical syndromes (e.g., paroxysmal cold shapes of degenerating RBCs and, importantly, recog-
hemoglobinuria [PCH], paroxysmal nocturnal hemo- nized that such cells were the origin of hemoglobin.4
globinuria [PNH], and march hemoglobinuria) char- Anemia without Blood Loss. In 1843, Andral
acterized by hemoglobin in the urine. (Fig. 1-2) described a spontaneous anemia, which
Investigators have not always been farseeing and arises without any prior blood loss.13 He quantified
logical, moving steadily and directly to their goal, nor red blood globules in healthy patients and reported
Historical Concepts of Immune Hemolytic Anemias 3

FIGURE 1-1. Antonj van Leeuwenhoek (1632–1723). (From


Wintrobe MM: Milestones on the path of progess. In: Wintrobe
MM (ed): Blood, Pure and Eloquent. New York: McGraw-Hill Book
Company, 1980:1–31.)

FIGURE 1-2. Gabriel Andral (1797–1876). (From Wintrobe MM:


Milestones on the path of progress. In: Wintrobe MM (ed): Blood,
Pure and Eloquent. New York: McGraw-Hill Book Company,
1980:1–31.)
4 Immune Hemolytic Anemias

16 early case of anemia. Although he provided no


other information concerning the patients’ condition,
what is important in relation to hemolytic anemia is
the observation of anemia without prior blood loss.
Hemoglobinuria. Vogel, in 1853,14 stated that the
matter in the urine is the same as that in the blood and
suggested that the matter in the urine consists of a
“decomposition of blood discs.” He suggested that
the degree of blood decomposition can readily be
ascertained by the degree of coloration in the urine,
and he indicated a connection between fevers, colored
urine, decomposition of blood discs, and anemia. This
represents one of the early examples of the association
between a decreased RBC count and the term anemia.
It also represents early evidence suggesting that
anemia may be secondary to infections.

RED BLOOD CELL AGGLUTINATION

The description of the phenomenon of RBC agglutina-


tion and its development as a tool in elucidating blood
groups took place in the last 30 years of the nineteenth
century in Germany and Austria, and were reviewed in
depth in 2002 by Hughes-Jones and Gardner.15 The dis-
coveries were largely the work of three people: Adolf
Creite, a medical student in Göttingen, Germany;
Leonard Landois, Director of the Physiological Institute FIGURE 1-3. Adolf Creite, about 1920. (From Hughes-Jones NC,
Gardner B: Red cell agglutination: The first description by Creite (1869)
at the University of Greifswald, Germany; and Karl and further observations made by Landois (1875) and Landsteiner
Landsteiner, working in the Pathological Anatomy (1901). Br J Haematol 2002;119:889–893.)
Institute in Vienna, Austria.15
Adolph Creite. Creite’s (Fig. 1-3) almost unknown
contribution was published in 1869 under the title
“Investigations concerning the properties of serum
proteins following intravenous injection.”16 His work fresh rabbit blood, then you observe under the micro-
is quite remarkable in that it showed that serum pro- scope that in the regions where the foreign serum
teins had the property of both “dissolving” and bring- mixes with the rabbit red cells, the cells suddenly flow
ing about “clustering” of red cells, that is, lysis and together in a peculiar way forming different shaped
agglutination in present-day terms, anticipating the drop-like clusters with irregular branches. I believed
discovery of antibodies by a quarter of a century. that I had found an explanation for the appearance of
Creite injected sera from calf, pig, dog, sheep, cat, blood in the urine, as it was possible that some blood
chicken, duck, and goat into rabbits. The first three cells had dissolved completely.”
had little or no effect on the recipient, but the sera of Leonard Landois. RBC agglutination and lysis were
the latter five almost always resulted in the appear- put on an even firmer basis by Landois, who pub-
ance of “blood-stained urine,” general malaise, and lished an extensive monograph on the subject of
death of the animal. He noted that the urine was free transfusion,17 which included a section describing his
of intact RBCs. He concluded that serum contains in vitro experiments. In his experiments, Landois was
agents that are able to dissolve red cells “directly.” He successful in demonstrating both lysis and agglutina-
performed additional experiments in which he tion. (It should be noted that the terms lysis and agglu-
removed protein from the serum before its injection tination were not in use until the end of the nineteenth
and observed that “all of the urine samples examined century. For lysis, both Creite and Landois used a
until the evening of the following day are normal.” German word meaning “dissolve”; for agglutination,
Accordingly, he concluded that the most likely words translatable as “accumulation,” “ball forma-
active ingredients were serum proteins, but added, tion,” or “sticky clumps” were used.) Landois also
“However, I cannot say how they function.” distinguished agglutination from rouleaux, for which
He also performed in vitro experiments and pro- he used the term, “like rolls of coins.”
vided a remarkably clear account of what is probably Landois added 4 to 5 mL of clear serum into a test
the first description of agglutination. He reported, “If tube and then added fresh defibrinated blood. He
you add blood serum from any of the animals with incubated the mixture at 37°C to 38°C or at room tem-
which I have carried out my experiments to a drop of perature and observed the initiation of the RBC lysis.
Historical Concepts of Immune Hemolytic Anemias 5

“Sooner or later the mixture becomes completely clear individuals.” More than a century later, his theories
and transparent and the cells are no longer visible. I about isoantigens are accepted and are a fundamental
observe the whole process of the lysis and the changes part of the theoretical basis of immunology, tissue
in red cell shape under the microscope.” Commenting transplantation, forensic medicine, and population
on another experiment on the mixing of cells and genetics.21,22
serum, Landois described the changes in shape of
RBCs and added, “The cells develop the ability to
stick to neighboring cells” and “form larger or smaller FIRST DESCRIPTION OF
clumps.” HEMOLYTIC ANEMIA
Karl Landsteiner. At the turn of the century, there
was a considerable amount of disagreement and con-
fusion about the occurrence and significance of The concept that premature destruction of RBCs
agglutination in both health and disease.15 It was at might lead to a disease state and jaundice was first
this point that Landsteiner (Fig. 1-4) entered the suggested in 1871 by Vanlair and Masius.1,23 These
field.17a,b The first suggestion of the existence of observers described a patient with anemia and
serum agglutinins and red cell antigens within what marked splenomegaly without hepatomegaly. The
would finally be known as the ABO blood group patient suffered acute attacks of left upper quadrant
system is to be found as a footnote in a publication by pain and jaundice without acholia, and passed
Landsteiner in 1900.18 In it he states, “The serum of reddish brown urine. Morphologic evidence of an
healthy individuals not only have an agglutinating RBC abnormality was suggested by finding spheri-
effect on animal red cells but also on human red cells cal dwarf cells in the peripheral blood that they
from different individuals. It remains to be decided called microcytes. The authors postulated that clini-
whether this phenomenon is due to individual differ- cal jaundice could result from two different mecha-
ences or to the influence of injuries or bacterial infec- nisms: “mechanically by reabsorption or liver
tion.” In a detailed paper in 1901, he reported that he induced” and “paradoxical icterus.” The latter
obtained sera and red cells from 29 different people, group included the “blood induced icterus,” where
including himself and four medical colleagues, to excessive amounts of colorant material is released
study agglutination reactions. The reason that from the blood cells and followed by the formation
Landsteiner was successful in elucidating the mecha- of bile which is deposited in the tissues. More explic-
nism underlying intraspecies agglutination where itly, they stated that “there are at least a certain
others had failed arose from the nature of number of non-mechanical types of icterus which
Landsteiner’s experimental design. He used all of the are caused by the exaggerated destruction of red
sera against all of the samples of RBCs, using cells and the transformation to bilirubin of released
“checkerboard” blocks of five or six different sera and hematin.” This concept was essentially correct, but
RBCs in 144 combinations. He found that certain sera little attention was paid to this remarkable publica-
would agglutinate the RBCs of certain other people. tion and, for almost 30 years, hepatic disease, jaun-
This discovery of isoagglutination became the basis dice, and hemolytic anemia became hopelessly
of human blood-group classification, which would intertwined.1
subsequently be found to have relevance for autoan-
tibody specificity in AIHA. THE DISTINCTION BETWEEN
In his characteristically brief but data-filled paper CONGENITAL AND ACQUIRED
of 1901,19 Landsteiner further noted and pointed out
that the blood isoagglutinins retained their activity HEMOLYTIC ANEMIAS
after drying and redissolving. Also, he observed
agglutination with serum extracted after 14 days At the turn of the twentieth century, Hayem24 (Fig. 1-6)
from blood dried on a cloth. “The reaction may be and Minkowski25 showed that the jaundice associated
suited to establish the identity or more correctly the with hemolytic anemia was distinct from that of
non-identity of a blood specimen.” This predicted hepatic diseases. Hayem made the distinction between
the value of Landsteiner’s discovery to forensic med- congenital and acquired hemolytic anemias, whereas
icine in the future. The closing statement in his paper Minkowski described only a hereditary condition.
was, “Finally, it might be mentioned that the Hayem has repeatedly been said to be the first to
reported observations may assist in the explanation describe acquired hemolytic anemia, although he did
of various consequences of therapeutical blood not name it that, but, instead, coined the term chronic
transfusions.” In three pages, Landsteiner com- infectious splenomegalic icterus.24 Minkowski is credited
pressed knowledge that would fill thousands of with the first clear recognition of icterus due to
pages in the future.20 hemolytic anemia (chronic hereditary acholuric icterus)
On November 8, 1930, Karl Landsteiner was separate from obstructive jaundice; he associated the
awarded the Nobel Prize (Fig. 1-5). The lecture given anemia with urobilinuria and splenomegaly and pos-
by Landsteiner at the conferment of his Noble Prize tulated that RBC destruction was attributable to lesions
was based on the “differences in the blood of human in the spleen.25
6 Immune Hemolytic Anemias

A B

C D
FIGURE 1-4. Karl Landsteiner at various times in his life. (A) Landsteiner at about the age of 5 (c. 1873), posing in a Husara riding costume on the pho-
tographer’s papier-maché rocks. (B) Photograph of Landsteiner probably taken at the Institute for Pathological Anatomy, where he worked from 1897

to 1907. (C) Landsteiner and his coworker, Emil Prásek from Belgrade, December 1913. The two worked together on the chemical manipulation of the
specificity of serum albumin. (D) Landsteiner at about the time he left Europe for the United States. (From Mazumbar MH: Species and Specificity. An
Interpretation of the History of Immunology. Cambridge, UK: Cambridge University Press, 1995.)
Historical Concepts of Immune Hemolytic Anemias 7

left upper quadrant abdominal pain, and spleno-


megaly shortly after giving birth. The patient’s
mother and sister were also icteric, and the sister’s
spleen was enlarged. The most remarkable aspect of
this paper lies in their description of the blood
findings. Although they made no mention of anemia
and had no concept of hemolysis as a pathological
process, they unmistakably described RBCs that we
now recognize as spherocytes with remarkable clarity
(Fig. 1-7). The authors noted that some of the RBCs,
which they called microcytes, were smaller than
normal RBCs, 3 to 4 μm in diameter, spherical in
FIGURE 1-5. The Noble Prize certificate for Karl Landsteiner in 1930. shape, and the contours were completely smooth.
(From Tagarelli A, Piro A, Lagonia P, Tagarelli G: Karl Landsteiner: A They concluded, “The jaundice of our patient appears
hundred years later. Transplantation 2001;72:3–7.) to be a peculiar type of icterus. The fact that the
patient’s mother and sister had a slight jaundice and
DESCRIPTION OF SPHEROCYTES AND that the sister had an enlarged spleen may indicate
that this condition is one disease entity.”
ANALYSIS OF THEIR SIGNIFICANCE Naegli is often credited with first use of the term
spherocyte. However, according to Crosby26 (Fig. 1-8),
Vanlair and Masius23 described the case of a young two British army officers, Christophers and Bentley,
woman who developed icterus, recurrent attacks of were the first. They were assigned to India to study

FIGURE 1-6. Georges Hayem. (From Packman CH: The


spherocytic haemolytic anaemias. Br J Haematol
2001;112:888–899.)
8 Immune Hemolytic Anemias

blackwater fever and made very careful descriptions of


spherocytes in a monograph published in 1909. Naegli
also proposed that the spherocyte was pathognomonic
of congenital hemolytic icterus, an observation that

FIGURE 1-8. William H. Crosby. (From Wintrobe MM: Blood, Pure and
Eloquent. New York: McGraw-Hill Book Company, 1980:XVIII.
Reproduced with permission of The McGraw-Hill Companies.)

constricted thinking about hemolytic icterus for the


next 15 or 20 years. In fact, many authorities began to
doubt the existence of an acquired type of hemolytic
icterus, regarding the disease as a variation on the con-
genital form.

OSMOTIC FRAGILITY OF RED


BLOOD CELLS

During the first decade of the twentieth century, a


number of significant studies of the osmotic fragility
of RBCs were conducted. Chaufford27 (Fig. 1-9)
noted that RBCs of several patients, but not those of
normal subjects, were hemolysed by hypotonic
saline. He developed an osmotic fragility test, in
which RBCs were placed in a series of tubes contain-
ing successively decreasing concentrations of saline.
The osmotic fragility was expressed as the concen-
tration of saline at which hemolysis began and at
which hemolysis was complete (Fig. 1-10). Chauffard
recognized that the liver was not at fault and that the
disorder was a result of hemolysis. He wrote,
“Perhaps after this clinical and hematologic inquiry,
FIGURE 1-7. A reproduction of part of the tinted lithograph illustrating the cause of the hemolytic theory could be consid-
the paper by Vanlair and Masius (1871) entitled De la micro- ered as won.” This observation finally enabled physi-
cythémie. I is a drawing of the patient’s blood. II is a drawing of control cians to distinguish hepatic and hemolytic jaundice,
normal blood. (From Dacie JV: The life span of the red blood cell and
circumstances of its premature death. In: Wintrobe MM (ed): Blood, as Ribbierre had recently (in 1903) demonstrated that
Pure and Eloquent. New York: McGraw-Hill Book Company, the cells from patients with hepatic jaundice are
1980:211–255.) resistant to osmotic stress.7
Historical Concepts of Immune Hemolytic Anemias 9

Of course, Chauffard and coworkers27 had discov-


ered the in vitro pathophysiological expression of the
spherical microcytes described by Vanlair and
Massius23 almost 40 years earlier. However, they
were probably unaware of the work of these early
investigators and they certainly made no association
between microcytic spherical cells and increased
osmotic fragility. That correlation was noted much
later by Haden.28

RETICULOCYTES

About 1 year after his description of increased


osmotic fragility in congenital hemolytic icterus,
Chauffard and Fiessinger29 and Chauffard30 stained
RBCs from patients with hemolytic icterus with
Pappenheim’s31 (Fig. 1-11) solution and noted large
numbers of cells containing a peculiar basophilic
granulation or reticulum, which they called “granu-
lar degeneration.” Ehrlich had first described this
special staining method in 18817 and noted increased
numbers of reticular cells in anemic patients.
Vaughan,32 in 1903, noted these granular cells consti-
tuted about 1% of the RBCs in normal subjects.
Chauffard had hoped to explain the anatomical lesion
that underlay the increased fragility of the RBCs.
What he actually discovered, or rediscovered, was the
reticulocytosis that is now a hallmark of hemolytic
FIGURE 1-9. Anatole Chauffard (1855–1932). (From Dacie JV: The life
span of the red blood cell and circumstances of its premature death.
anemia. Chauffard’s drawing30 of a blood smear
In: Wintrobe MM (ed): Blood, Pure and Eloquent. New York: McGraw- stained with Pappenheim stain from a patient with
Hill Book Company, 1980:211–255.) familial hemolytic icterus is shown in Figure 1-12.

Diamètre moyen des hématics 5 μ 89 Diamètre maxima 7,5.


— minima 4.
Hémolyse totale....

Hémolyse très nette

Hémolyse nette....

Hémolyse légère....

Pas d’hémolyse....
Nombre de gouttes de la
solution.... 70 68 66 64 62 60 58 56 54 52 50 48 46 44 42 40 38 36 34 32

Résistance globulaire — (Solution de NaCI à 0.70%)

FIGURE 1-10. The figure illustrates the “precocious and prolonged” lysis in hypotonic saline of the red cells of a patient suffering from ictère con-
génital de l’adulte (hereditary spherocytosis). (From Dacie JV: The life span of the red blood cell and circumstances of its premature death. In:
Wintrobe MM (ed): Blood, Pure and Eloquent. New York: McGraw-Hill Book Company, 1980:211–255.)
10 Immune Hemolytic Anemias

FIGURE 1-11. Artur Pappenheim (1870–1916). (From Lajtha LG: The common ancestral cell. In: Wintrobe MM (ed): Blood, Pure and Eloquent.
McGraw-Hill Book Company, 1980:81–95. Reproduced with permission of The McGraw-Hill Companies.)

THE CONCEPTS OF IMMUNE HEMOLYSIS thermolabile substance (variously termed comple-


AND HORROR AUTOTOXICUS ment, alexin, or cytase) to cause the specific destruc-
tion in vitro of the erythrocyte species used for
immunization.8 During the course of these studies,
In an impressive series of studies commencing in Ehrlich and Morgenroth attempted repeatedly to
1899,33 Paul Ehrlich (Fig. 1-13) and Julius induce the animal to form hemolytic antibodies to its
Morgenroth sought to identify the constituents and own cells. These attempts to elicit the formation of
to define the mechanisms involved in the phenome- autoantibodies were uniformly unsuccessful, and, at
non of immune hemolysis, which Jules Bordet had best, they were only able to produce antibodies able
only recently described.34 Such studies involved the to agglutinate or to hemolyse the RBCs of certain
immunization of animals with foreign RBCs, a pro- other members of the same species.
cedure resulting in an immune serum whose ther- Ehrlich had postulated, in his landmark paper of
mostable antibody would collaborate with a 1897, that antibody formation was part of the normal
Historical Concepts of Immune Hemolytic Anemias 11

In the latter part of the nineteenth century, there were


a number of reports of PCH. Dressler41 is generally
credited with being the first (in 1854) to give a clear
description. His patient was a 10-year-old boy who
may have had congenital syphilis. After exposure to
cold, he passed red urine that gradually paled to clear
to a natural color. Microscopic examination of the
urine showed “dirty brown pigment” but no blood
corpuscles. PCH, however, seems also likely to have
been the diagnosis in the patient described by
Elliotson in The Lancet in 18323,42 who had heart
disease and cold “fits” and passed bloody urine
“whenever the east wind blew.”
Subsequently, several excellent clinical accounts
were published during the 1860s.3 The authors real-
ized that exposure to cold precipitated that attacks
and that the urine contained blood pigment, but no
blood cells. Wiltshire43 described an infant, perhaps
the youngest such patient ever recorded, who passed
bloody urine, free from RBCs in the sediment, when
the “weather was particularly inclement.”
The term hemoglobinuria seems to have been used
FIGURE 1-12. Drawing of a blood smear (Pappenheim stain) as seen first by Secchi in 1872, but it is not clear whether the
by Chauffard (1908). The granular appearing cells are reticulocytes
from a patient with familial haemolytic icterus. (From Packman CH: The patient he described had PCH.44
spherocytic haemolytic anaemias. Br J Haematol 2001; In 1879, Stephen Mackenzie, at the London
112:888–899.) Hospital, elaborated on the pathophysiology of
PCH.45 He described a young boy who had a sallow
physiological process of cellular digestion and so complexion and yellow eyes and whose urine was
might theoretically be stimulated by autochthonous black. The microscopic examination and spectro-
as well as by foreign substances.8 Nevertheless, he scopic analysis of the urine showed it contained
pointed out, “It would be dysteleologic in the highest abundant hemoglobin but no RBCs. He suggested
degree, if under these circumstances self-poisons of that the discolored urine was due to blood solution or
the parenchyma⎯autotoxins⎯were formed.”35,35a disintegration (hemolysis) and stated that it must
Thus, “we might be justified in speaking of a horror take place in some part of the organism. He believed
autotoxicus of the organism.”36 that the hemolysis occurred in the “genito-urinary
apparatus,” most probably the kidney.
Kuessner, in 1879, made the important observation
THE FIRST DESCRIPTION OF AN that serum obtained by “cupping” a patient during an
AUTOIMMUNE HEMOLYTIC ANEMIA attack of hemoglobinuria was tinged red.46 This prob-
ably was the first direct evidence derived from obser-
vations in humans that indicated that the hemoglobin
The first AIHA in which clinical and diagnostic labora-
in the urine was being derived from hemoglobin
tory findings were clearly described is PCH.37 This
liberated in the plasma, rather than being, in some
appears, at first, to be surprising because PCH is the
mysterious way, of renal origin. Indeed, Mackenzie
least common type of AIHA. Its early recognition is due
modified his previous theory of erythrocyte destruc-
to the fact that hemoglobinuria is a striking symptom,
tion, suggesting that the role of the kidney is in fact
a fact that also explains the early recognition of march
passive, and that the corpuscle solution, or hemolysis,
hemoglobinuria and PNH. It is also true that PCH was
occurs in the vasculature.47
much more common than it is at present because a
majority of cases recorded in the early medical litera-
ture were associated with late stage syphilis or congen- EARLY DIAGNOSTIC TESTS FOR
ital syphilis. In the early 1900s, over 90% of patients
with chronic PCH had a positive test for syphilis and
PAROXYSMAL COLD HEMOGLOBINURIA
approximately 30% showed clinical evidence of the
disease.38 With the effective treatment of syphilis and Although there were many clinical descriptions of
the virtual elimination of the congenital form, “classic” PCH in the nineteenth-century medical literature
syphilitic PCH is now an extremely rare disorder, as is documenting the relationship of acute attacks to expo-
chronic PCH. It was in patients with the chronic form sure to cold and the fact that the urine contained
of PCH that exposure to cold resulted in a paroxysm of blood pigment but no blood corpuscles, the patho-
hemoglobinuria.39,40 physiology was not understood.
12 Immune Hemolytic Anemias

FIGURE 1-13. Paul Ehrlich (1854–1915) in his


study.* (From Wintrobe MM: Milestones on
the path of progress. In: Wintrobe MM: (ed):
Blood, Pure and Eloquent. New York: McGraw-
Hill Book Company, 1980:1–31.)

A diagnostic test described in 1879 was based on the temperatures and that labile serum factors (comple-
development of hemoglobinuria after immersion of ment) caused lysis of the sensitized cells if the temper-
the patient’s feet in ice water.39,40 A test producing less ature was subsequently raised. Their interpretation of
discomfort to the patient was described in 1881 by their observations are particularly noteworthy because
Ehrlich, who showed that if a ligature was placed they were published during the era of widespread
around a finger that was then chilled in ice water, acceptance of Ehrlich’s dictum of horror autotoxicus.
serum subsequently obtained from the finger would Here then was the first report that appeared to contra-
contain hemoglobin.48 Although these tests helped to dict Ehrlich’s concept.35a
diagnose the disorder, they did not elucidate the This bithermic procedure for the diagnosis of PCH
mechanism by which exposure to cold resulted in was the first immunohematologic test ever to be
hemolysis. described51 and remains the diagnostic test for the dis-
order (see Chapter 5). Further, this discovery has been
widely acclaimed as the first description of an autoan-
THE DONATH-LANDSTEINER DISCOVERY, tibody and of an autoimmune human disease.8 The
1904: THE FIRST DESCRIPTION OF AN test is referred to as the Donath-Landsteiner (DL) test
AUTOANTIBODY AND OF AN and the antibody thus detected as the DL antibody.
AUTOIMMUNE HUMAN DISEASE Even after the passage of a century since the report of
Donath and Landsteiner, the accuracy of their obser-
vations and the usefulness of the DL test persist.
The greatest single step forward in understanding the Primacy of Discovery of Biphasic Autoantibodies
pathogenesis of PCH was provided by the work of in Paroxysmal Cold Hemoglobinuria. It is of interest
Donath and Landsteiner whose famous report was that similar and apparently independent observations
published in 1904.49 Julius Donath (1970–1950) was an were described by Eason. Easons’s two papers,52,53
assistant at the University of Vienna First Medical published in 1906, were based on his MD thesis. His
Clinic, and Karl Landsteiner (1868–1943) became a experiments, which had been carried out in 1903, had
giant in the annals of immunology.50 These investiga- been the subject of a communication read at a meeting
tors demonstrated that hemolysis was due to an of the Galenian Society, Edinburgh, in January 1904. He
autolysin that reacted with the patient’s RBCs at low stated that “ten months after the results had been
communicated by me the most important of them were
confirmed by Donath and Landsteiner whose research
on these lines had been conceived independently of
*This is the authors’ favorite photograph, indicating that offices
circa 1899 were not necessarily neater than those of the present
mine. These collaborators furthermore proved that it is
day. It certainly seems as though Ehrlich maintained enough the process of anchoring of the intermediary body to the
reading material in his office. red corpuscles which requires the low temperature.”
Historical Concepts of Immune Hemolytic Anemias 13

Dr. Eason was awarded a Gold Medal and the Milner- Their article describes further experiments in which
Fothergill Medal in Therapeutics by Edinburgh two aliquots of a patient’s blood were obtained. One
University for his thesis.37 aliquot was incubated at 0°C, and the other was incu-
However, Donath and Landsteiner contested the bated at room temperature. Then the plasmas were
priority for their discovery with Eason and stated that removed and exchanged, mixed, and incubated at
“Eason joined [himself] to our interpretation of the 37°C. After 2 hours, the aliquots that were cooled had
mechanism of hemolysis.“8,54 They further stated that undergone much lysis, but no lysis occurred in the
“the development of autotoxic substances, which are other aliquot. “This finding indicates that red cells
bound to the organism’s own cells, can be related to the take up in the cold an effective substance from the
process of antibody formation, a possibility which, so plasma, and that neither red cells nor white blood
far as we know, has not previously been discussed.”8,54 cells give hemolytic substance into the serum.”
In a much more recent publication, Goltz55 main- In an additional experiment, “oxylated blood” of
tained that Donath and Landsteiner did not actually the patient was cooled in ice water and centrifuged in
discover the first autoantibody because nowhere did the cold, and then the plasma that had been removed
they use the accepted terms “antibody,” “ambozep- in the cold was mixed with a new aliquot of red cells
tor,” “antigen,” or even “immune.” Rather they used of the patient. This mixture was then cooled and sub-
such apparently nonspecific terms as “hemolysin,” sequently incubated at 37°C. However, no hemolysis
“toxin,” and “poison.” However, as reviewed in depth occurred, thus indicating that the hemolysin had been
by Silverstein,8 numerous contemporary authors used absorbed by the cells.
the term toxin when they meant “specific antibody,” “Red cells that are cooled with serum or plasma of
and the term did not imply some sort of nonimmuno- hemoglobinuric patients, whether the patient’s own or
logic toxic action. other’s red cells, take up substances that by this absorp-
Even if Landsteiner’s language might be misinter- tion develop the capability to hemolyse in the serum of
preted at a later period, his contemporaries surely hemoglobinuric patients and other human serum. The
understood him. For example, Ehrlich in 1906 already hemolysis is caused by the aid of factors in the serum
referred to Donath and Landsteiner as observing described as complement (alexin, cytase, etc.).”
“hemolytic autoamboceptors.”8,33 Further, Rössle, in
1909,8,56 while discussing the general evidence of the
existence of autoantibodies, stated that “there are also FURTHER STUDIES ON THE
cases, however, in which direct evidence for the pres- MECHANISMS OF HEMOLYTIC ANEMIA
ence of autoamboceptor is splendid. The best known AND OBSERVATIONS ON THE
instance concerns paroxysmal hemoglobinuria.” “Even DISTINCTION BETWEEN CONGENITAL
in their first report, Donath and Landsteiner called our
attention to the possibility that such a substance might AND ACQUIRED FORMS
be the result of self-immunization.” Also in 1909,
Meyer and Emmerich published an extensive report on Chauffard was among several French scientists who
paroxysmal hemoglobinuria.57 They concluded their explored the mechanisms of hemolytic anemia in the
paper with the statement that “In [our] four cases of early years of the twentieth century.4 Chauffard
typical paroxysmal cold hemoglobinuria, the autohe- (1907),27 along with Trosier (1908)59 and Vincent
molysin found by Donath and Landsteiner was (1909),60 described autohemolysins in patients with
observed.” acute acquired hemolytic jaundice. These authors
It is evident from the foregoing that Donath and described patients whose serum had the capability of
Landsteiner as well as their contemporaries did, hemolysing RBC, and they termed the condition
indeed, understand from the outset that they were “hemolytic icterus”; it was acute in course and associ-
describing an autoantibody and an immunological ated with hemoglobinuria. The reports of hemolysins,
process, despite the curious terminology they used.8 although incomplete and to some extent unsatisfac-
The Original Experimental Protocol of Donath tory, were pioneer ones well in advance of their time,
and Landsteiner. Excerpts from the original report in and the idea that hemolytic anemia could occur appar-
1904 by Donath and Landsteiner58 are illustrated in ently spontaneously in humans in consequence of the
Figures 1-14 and 1-15. A translation of the original development of abnormal agglutinins or hemolysins
protocol is provided in Figure 1-16. remained controversial for the next 30 years or so.3
In essence, they demonstrated that sera from Chauffard also standardized the osmotic fragility
patients with PCH would cause hemolysis in vitro of test, described reticulocytes and their increased
RBC of normal individuals and of patients if the numbers in congenital hemolytic icterus (later to be
serum and cells were held (incubated) for 1⁄2 hour at known as hereditary spherocytosis), and drew atten-
5°C and then held at 37°C. As controls, they used tion to the microcytic nature of the RBCs in some
serum from normal individuals. They concluded that hemolytic anemias.27
the serum of the hemoglobinuric patients contains a Between 1908 and 1912, Widal, Abrami, and
lytic substance that is effective against the patients’ Brule61,62 introduced the term acquired hemolytic
and other human blood corpuscles. anemia. These investigators described hemolytic
14 Immune Hemolytic Anemias

FIGURE 1-14. The original report published in 1904 by Dr. Julius


Donath and Dr. Karl Landsteiner describing their current theories of
the pathogenesis of paroxysmal cold hemoglobinuria and the develop-
ment of the biphasic lysis test that remains the diagnostic laboratory
procedure for the disorder. A translation of portions of the text follows
(a more complete translation has been published by Bibel50).

About Paroxysmal Hemoglobinuria


Different theories have been proposed to explain the pathogenesis
of paroxysmal hemoglobinuria, a peculiar illness whose attack under
the influence of cold leads to hemoglobinuria and removal of blood
pigment through the urine.
Other, older explanations state that hemoglobinuria is caused by FIGURE 1-15. The original protocol of the experiments performed by
the destruction of blood corpuscles in the kidney. But after Küessner Donath and Landsteiner and their interpretation. A translation of the
showed that hemoglobinemia is present during such paroxysms, the protocol is given in Figure 1-16. (From Donath J, Landsteiner K: Uber
cause was located in the blood. The hemolysis itself was thought to be paroxysmale Haemoglobinurie. Munchen Med Wschr 1904;51:1590.)
dependent on various factors. The original belief that cold would
destroy the red cells that are sensitive in this disease is in opposition
to the commonly acknowledged fact that the blood of these patients in
vitro is not more sensitive to cold than the blood of normal individuals. Hayem 10 years earlier. The patients exhibited
Therefore one had to look for other causes of the hemolysis. Recent reticulocytosis, but the alterations in the fragility test
extensive studies on blood toxins have suggested that this disease is were less marked than in the congenital form.
caused by hemolysins. Authors have spoken for the hemolytic effect of Hence, at this time, the two types of hemolytic
these toxic substances. But numerous efforts to find the toxic agents
did not succeed exactly, or even to find a test system that allows one
anemia were well defined: the congenital form of
to study the hemolytic procedure during the period of hemolysis. (From Minkowski and Chauffard and the acquired form of
Donath J, Landsteiner K: Uber paroxysmale Haemoglobinurie. Hayem and Widal (Figs. 1-6 and 1-17).
Munchen Med Wschr 1904;51:1590.)

icterus that was apparently neither congenital nor


THE ROLE OF THE SPLEEN AND
familial, that could appear gradually or suddenly THE EFFECT OF SPLENECTOMY
during the course of various diseases, or that could
be unassociated with any underlying disease. These The above-cited brilliant studies clearly distinguished
cases were considered similar to those described by hepatic jaundice and the jaundice resulting from
Historical Concepts of Immune Hemolytic Anemias 15

Held for 1/2 hr not generally regarded as a significant site of RBC


Blood Cells at 5°, then 21/2 Held 3 hours destruction. Therefore, it is not surprising that in 1911,
Serum 3 Drops hr at 37° at 37°
Micheli, in Turin, performed the first splenectomy for
Patient K Patient K Ruby red 0
acquired hemolytic anemia.64 Banti65 (Fig. 1-18),66 in
(hemoglo- B.W. Red 0 1912, conducted an extensive investigation into splenic
binuria) Ch.G. Red 0 pathology and introduced the term hemolytic spleno-
4 Drops A.R. Red 0 megaly, when he observed that the spleens of animals
undergoing hemolysis were enlarged and congested.65
Patient R Patient R Ruby red Trace of red He also noted that heteroimmune hemolytic serum,
(hemoglo- B.W. Ruby red Trace of red
when transfused into splenectomized animals, led to
binuria) Ch.G. Ruby red Trace of red
10 Drops A.R. Ruby red Trace of red
less and slower hemolysis than that seen with normal
animals. He implicated the splenic endothelial cells as
Patient N Patient N Ruby red 0 erythrophagocytes and described agglutinated erythro-
(hemoglo- B.W. Ruby red 0 cytes within the splenic pulp. Banti similarly showed
binuria) Ch.G. Red 0 that the Kupffer cells of the liver could have an ery-
7 Drops A.R. Ruby red 0 throphagocytic function when intense hemolysis was
present.
B.W. B.W. 0 0
6 Drops Patient R 0 0
Thus, Banti effectively described the reticuloendo-
Patient N 0 Trace of red theilal system and its function in RBC hemolysis.4 He
Ch.G. Red tinged Clear distinct red recognized the importance of the spleen to the disease,
but stressed that it was not the only, nor even the prime,
Ch.G. Ch.G. 0 0 site of RBC destruction. The combined activities of
7 Drops Patient K 0 0 Micheli and Banti entrenched the recommendation of
Patient N 0 0 splenectomy as a treatment for hemolytic anemia, rep-
A.R. Patient K 0 0
resenting the first specific therapy for AIHA. Despite
6 Drops Patient N 0 0 the widespread acceptance of the benefits of splenec-
Patient R 0 0 tomy, however, some, such as Antonelli, in 1913 refuted
B.W. 0 0 Banti’s hemolytic splenomegaly as a separate disease,
Ch.G. 0 0 pointing out that it did not differ from acquired
hemolytic anemia.4
FIGURE 1-16. It is shown with this sequence of experiments that the
blood corpuscles of other individuals are hemolyzed by the serum of
patients with hemoglobinuria, although to a lesser degree than their FURTHER CHARACTERIZATION
own blood corpuscles; however, in the same series of experiments,
the blood corpuscles of the hemoglobinuric patients which have been
OF HEMOLYTIC ANEMIAS
cooled with other serum do not lyse when they are warmed after-
wards. (Serum B. W. had a normal isolytic activity against the red World War I brought a halt to investigation and case
cells of Ch.G. and N which was not increased by cooling.) Therefore,
the unusual composition of the blood of the hemoglobinuric patients
reports of hemolytic icterus.7 By the 1920s, the prevail-
which is causing the lysis lies in the serum (respectively plasma), ing understanding of the mechanism behind RBC
although the red cells may be easier to lyse (as shown in our Case destruction was that it resulted from autoagglutinin-
K). The serum (plasma) of the hemoglobinuric patients contains a induced agglutination, the first step in hemolysis.
lytic substance that is effective against the patient’s and other However, publications after World War I indicated the
human blood corpuscles. This lysis cannot be demonstrated directly
by mixing the serum of the hemoglobinuric patient with his own or degree to which much of the knowledge discovered at
other red cells; however, one must consider the dependence of its the beginning of the century had been lost. Lederer
effects on temperature. (1925)67,68 and Brill (1926)69 described a number of cases
of transfusion-responsive acute hemolytic anemia asso-
ciated with infectious diseases. Because much of the
premature and excessive destruction of erythrocytes. prior French work had been forgotten, Lederer’s
The hemolytic process was further differentiated to descriptions were thought to be of a new disease, in
include both congenital and acquired forms. Although spite of the extensive review of hemolytic icterus by
the phenomenon of agglutination had been well Tileston70 just 3 years earlier. Such cases became known
described in the latter part of the nineteenth century, it as “Lederer’s anemia” or “Lederer-Brill anemia,” but it
was Widal, Abrami, and Brulé who observed autoag- is likely that they were examples of AIHA.1
glutination of erythrocytes,4 and their work as well as The hiatus in studies concerning hemolytic anemias
others was summarized at the twelfth session of the obliterated the clear distinction between congenital and
Congrès Français de Medicina, which took place in acquired forms of hemolytic anemia established by the
Lyon in 1911.63 The topic was the role of hemolysins in French investigators. Indeed, Dacie71 states that it was
pathology, and papers were presented by many of the generally assumed at that time in England that
foremost physicians and pathologists of the day. hemolytic anemia occurring in the adult was a latent
By now the role of the spleen was widely accepted form of hereditary spherocytosis. The lack of specific
as being the major site of hemolysis, and the liver was diagnostic procedures, the presence of spherocytes in
16 Immune Hemolytic Anemias

FIGURE 1-17. Fernand Widal. (From Packman CH: The


spherocytic haemolytic anaemias. Br J Haematol
2001;112:888–899.)

both forms of hemolytic anemia and the unavailability acquired hemolytic icterus in 1940 that was 96 pages
of serologic testing made such a conclusion inevitable.1 in length with 380 references. They identified 81 arti-
In 1938 and 1940, important contributions were cles that described cases fitting their concept of acute
made by Dameshek (Fig. 1-19) and Schwartz.72-74 (acquired) hemolytic icterus. Based on their own
These workers published a remarkable review of clinical observations of hemolysins in some patients,

FIGURE 1-18. Guido Banti (1852–1925) was one of the first physicians who might prop-
erly be called a hematologist. A contemporary of Osler, he worked at a time when the
methods and laws of biological research were just developing. Medical discovery was com-
monly a consequence of clinical insight aided only by physical examinations and necropsy.
The titles of Banti’s earliest publications give the direction of his lifelong interests: “Splenic
anemia” and “Enlargement of the spleen with cirrhosis of the liver.” His efforts to define
these conditions as entities came to nothing, but the discussions about them did much to
demonstrate the essentialness of method in clinical research. (From Crosby WH: The
spleen. In: Wintrobe MM (ed): Blood, Pure and Eloquent. New York: McGraw-Hill Book
Company, 1980:97–138. Reproduced with permission of The McGraw-Hill Companies.)
Historical Concepts of Immune Hemolytic Anemias 17

FIGURE 1-19. William Dameshek (1900–1969), one of the


most eminent of American hematologists of his era, was a
strong proponent of the concept of autoimmunity at a time that
others were reluctant to accept that a patient could produce
autoantibodies. His extensive writings and teachings had a
major influence on the gradual acceptance of an autoimmune
etiology for some types of acquired hemolytic anemias. (From
Crosby WH: The Spleen. In: Wintrobe MM (ed): Blood, Pure and
Eloquent. New York: McGraw-Hill Book Company, 1980:97–138.
Reproduced with permission of The McGraw-Hill Companies.)

cases reported in the literature, including those of Thereafter, during subsequent decades, the
Chauffard and coworkers, and their own experi- classification and serological characteristics of the
ments involving injection of varying amounts of various AIHAs were delineated, in large part through
hemolytic serum into guinea pigs, they proposed the extensive and meticulous work of Sir John Dacie
that all cases of hemolytic icterus were a result of in London.5,74a
hemolysins. The differences in clinical manifesta-
tions, ranging from mild congenital cases to fulmi- MEASUREMENTS OF RED
nant acute hemoglobinurias, were accounted for by
the dose of hemolysin.7 Dameshek and Schwartz’s BLOOD CELL SURVIVAL
general thesis that hemolysins were responsible for
the development of many cases of acquired In a review in 1923, Payton Rous (Fig. 1-20) discussed
hemolytic anemia was correct. However, they were the question of whether the RBCs had a definite, as
incorrect in extrapolating their concept of the role of opposed to an almost indefinite, sojourn in the blood,
hemolysins to congenital hemolytic jaundice (hered- and, if finite, how long was their life span.75 In fact, he
itary spherocytosis) and in concluding that that dis- did not doubt that their life span was limited, and he
order might be caused by the “more or less listed a number of cogent arguments in favor of this
continued action of an hemolysin.” view. For example, he cited the “continuous activity of
These studies reawakened interest in acquired broadly distributed hematopoietic tissue” and the
hemolytic anemia and laid the broad outline for our “daily excretion through the bile of a pigment nearly
modern concepts of the clinical and serologic implica- if not precisely identical with one of the pigmented
tions of AIHA.1 However, the difficulty in ascribing derivatives of hemoglobin.”75 The question as to how
cases of acquired hemolytic anemia to the develop- long RBCs circulate before undergoing destruction
ment of “hemolysins” was that they could not be had been a vexing question for many years. A variety
demonstrated in the vast majority of cases by the sero- of methods and calculations had been used to come
logic techniques then available. up with some answers, ranging from observations of
18 Immune Hemolytic Anemias

FIGURE 1-20. Peyton Rous (1879–1970). (From Dacie JV: The life span of the red blood cell and circumstances of its premature death. In:
Wintrobe MM (ed): Blood, Pure and Eloquent. New York: McGraw-Hill Book Company, 1980:211–255.)

the time it took for the RBC count in a hypertrans- at the time) but to the functioning of the transfused
fused animal to be restored to normal to calculations RBCs. By 1921, Ashby78 was able to report on more
based on bile excretion. The conclusions drawn from than 100 patients. In four patients who were followed
these studies were, however, erroneous. until the elimination of the transfused RBCs was com-
Data of Winifred Ashby. The conclusions of only one plete or almost complete, this did not take place until
observer stood out in striking contrast to the above 83 to 100 days after transfusion.
observations⎯those of Winifred Ashby (Fig. 1-21), One of the difficulties inherent in Ashby’s work,
whose first papers76,77 were published in 1919 (re- which she could not circumvent, was that she was not
viewed by Dacie3). Ashby described in her first paper measuring the life span of the RBCs in their own envi-
how she had transfused group IV (type O) blood into ronment. This raised the question of whether the
seven group II (type A) recipients who were suffering foreign cells might have a different survival than those
from various anemias and how she had been able of the host, a point that she was unable to resolve.
to count the free (unagglutinated) type O RBCs by Additional Studies Using Differential Aggluti-
making suspensions of posttransfusion blood in an nation. In 1928 differential agglutination was also
anti-A serum (Fig. 1-22). She concluded that transfused used in the reverse way by Landsteiner, Levine
RBCs live a long time, 30 days or longer, and that the (Fig. 1-23) and Janes79 and Wiener.80 Wiener reported
beneficial results of blood transfusion are not due to the that he had detected blood group M (or N) cells, using
stimulation of the bone marrow (a view held by some anti-M (or anti-N) sera, in the circulation of N (or M)
Historical Concepts of Immune Hemolytic Anemias 19

FIGURE 1-21. Winifred Ashby (1879–1975). (From Dacie JV: The life
span of the red blood cell and circumstances of its premature death.
In: Wintrobe MM (ed): Blood, Pure and Eloquent. New York: McGraw-
Hill Book Company, 1980:211–255.)

recipients for between 80 and 120 days after transfu-


sion. Wiener also used the Ashby method, using anti-
M (or anti-N) sera to agglutinate the recipient’s RBCs,
and observed that between one third and one fourth FIGURE 1-22. Reproduction of one of Ashby’s original figures. (A) A
of the transfused RBCs disappeared each month; he suspension of group II (type A) red cells in an anti-A serum. Relatively
remarked that this continuous decrease in numbers few cells are free and unagglutinated. (B) A similar preparation after
was to be expected on the assumption that all the the transfusion of group IV (type O) red cells. Many of the cells are now
free and unagglutinated, the great majority being transfused cells.
cells had approximately the same life span. He con- (From Dacie JV: The life span of the red blood cell and circumstances
cluded, “Curiously enough, despite all this work, of its premature death. In: Wintrobe MM (ed): Blood, Pure and
most textbooks still give the life of the erythrocyte as Eloquent. New York: McGraw-Hill Book Company, 1980:211–255.)
thirty days.”
Ashby’s data and conclusions are now known to be
generally correct. But she was ahead of her time; her hemolytic anemia over 20 years after her publication.
papers remained on library shelves largely unread They were able to show that normal RBCs transfused
and her technique was relatively unused until the late into patients with familial hemolytic anemia sur-
1930s. In Oslo, Dedichen81 conceived the idea that it vived normally, for approximately 100 to 120 days.
might be possible to obtain evidence by transfusion The survival curves from their paper are shown in
experiments as to which of the two current theories Figure 1-26. In sharp contrast, Loutit and Mollison83
about the pathogenesis of “ictere hemolytique” noted that normal RBCs transfused into patients
(hereditary spherocytosis) was correct; hyperactivity with acquired hemolytic anemia exhibited markedly
of the organs of hemolysis or production of cells with reduced survival.
less than normal resistance. However, for technical Loutit and Mollison83 also transfused RBCs from
reasons, his experiments were unsuccessful, and more patients with congenital and acquired hemolytic icterus
than a decade was to pass before similar (but more into normal recipients and followed their survival. The
successful and decisive) experiments were again RBCs from patients with congenital acholuric jaundice,
undertaken. including those from a patient who had undergone
Intrinsic and Extrinsic Mechanisms of Hemo- splenectomy, exhibited short survival.
lysis. Dacie (Fig. 1-24) and Mollison (Fig. 1-25)82 The tracing by differential agglutination, as intro-
first applied Ashby’s technique in patients with duced by Ashby, demonstrated a clear distinction
20 Immune Hemolytic Anemias

FIGURE 1-23. Philip Levine. (From Diamond LK: The story of


our blood groups. In: Wintrobe MM (ed): Blood, Pure and
Eloquent. New York: McGraw-Hill Book Company,
1980:691–717. Reproduced with permission of
The McGraw-Hill Companies.)

between the two major groups of cases. In one group, serious disease in horses and humans and for which
transfused blood survived normally, and in another there was no cure at that time. He later continued his
group of patients, it was destroyed along with the work at Cambridge in the University Department of
patient’s own blood. These observations supported Pathology. Two eminent serologists, Robert Race (see
the idea that there might be “intrinsic” and “extrinsic” Fig. 1-25B) and Arthur Mourant, were working in the
mechanisms for increased hemolysis. Later, the dis- department at that time. Race86 and Weiner,87 working
tinction was used as a rational basis for classification separately, had by this time concluded that there were
of the hemolytic anemias. two types of Rh antibody: one that bound to the RBC
surface and caused agglutination (the “complete”
antibody) and another that absorbed to the RBC
THE ANTIGLOBULIN (COOMBS’) TEST surface but did not cause agglutination (the “incom-
plete” antibody). Coombs, reminiscing in 1998,84
A major diagnostic advance was the development of states, “At coffee one day, discussion turned to Rob’s
the antiglobulin test, the discovery of which is an so-called ‘blocking’ or ‘incomplete’ antibody. What
interesting aspect of the history of AIHA. The events was the nature of this antibody, if indeed it was an
leading to its discovery have been documented by Dr. antibody? Rob stressed that there was a real need for
Robin R. A. Coombs84,85 (Fig. 1-27). a better test (than his blocking test) to measure these
He points out that immunology in the 1940s was so-called incomplete antibodies. The next step
somewhat elementary, unsophisticated, and pheno- occurred on a late-night ill-lit train from London back
menologic. The real nature of antibodies was still to Cambridge. I was pondering on how to measure
uncertain, but seemed to be associated with the serum these incomplete antibodies on red cells with pictures
globulins. After graduating in veterinary medicine in in my head of Ehrlich’s side-chain theory. In a flash I
1943, he joined an investigation on the serodiagnosis could see the globulin antibody on the red cells, and
of Pfeifferela mallei infection, which causes a very these red cells should be agglutinated with an anti-
Historical Concepts of Immune Hemolytic Anemias 21

added to the proofs as an addendum. Coombs states,


“The lesson is that one should never refer to a discov-
ery or a test as being new.”84
Coombs, Mourant, and Race next went on to demon-
strate RBC sensitization in babies with hemolytic
disease of the newborn using the direct antiglobulin
test (DAT).91 Cord RBCs from patients agglutinated
when exposed to the antihuman antiglobulin reagent,
but cells from healthy babies did not agglutinate.
One of the positive tests they observed in newborns
appeared at first to be a false positive since there were
no Rh antibodies in the mother’s serum. However,
Race went on to demonstrate the test was a true posi-
tive but that it was not caused by an Rh antibody. The
mother’s name was Kell, and this was the start of
Race’s research on the Kell blood group system.
In 1947, Coombs and Mourant92 demonstrated that
the component in AGS that reacted with RBCs coated
with Rh antibody was in all probability an anti-
gamma globulin. They showed that the addition of a
small amount of gamma globulin to the antiglobulin
serum rendered it incapable of agglutinating cells
FIGURE 1-24. Professor Sir John Dacie laid the foundation for the coated with Rh antibody, whereas the addition of
investigation of hemolytic anemias. His persistence and experimental alpha globulin or beta globulin had only a slight
approach enabled him to demonstrate the vast complexity of the effect, which could be ascribed to contamination with
factors involved in the anemias due to hemolysis, and for this he has traces of gamma globlulin.
justifiably been considered a pioneer.81a He was also responsible for
training many hematologists from numerous countries, including the An interesting phenomenon observed by Dacie93
present authors. (From Wintrobe MM: Blood, Pure and Eloquent. New was that the addition of gamma globulin to AGS
York: McGraw-Hill Book Company, 1980:XVIII. Reproduced with produced a reagent that could discriminate between
permission of The McGraw-Hill Companies.) the RBCs of individual patients with AIHA. Thus,
although in many instances the positive antiglobulin
body to serum globulin, i.e., an antiglobulin. All the reaction was abolished by adding the gamma globulin,
necessary thinking had been done!” this was not true in all cases. It seemed clear that in
Coombs obtained some “very crude [rabbit] anti- those cases in which the reaction was inhibited, the
human globulin serum” from a coworker and the “very autoantibody on the cell was itself a gamma globulin,
first experimental protocols with Race and Mourant but that when the reaction was not affected, the mate-
showed quite clearly that the procedure was going to rial on the RBC surface could not be gamma globulin.
work.” They absorbed the antiglobulin serum (AGS) The “nongamma protein” was eventually shown to
with human group AB Rh-positive RBCs and then consist of components of complement fixed to the cell
incubated Rh-positive RBCs in sera known to contain as a result of antibody-antigen interaction.93,94
incomplete Rh antibodies. The sensitized cells aggluti- Use of the Antiglobulin Test to Distinguish Immune
nated in the antiglobulin serum and the appropriate from Nonimmune Acquired Hemolytic Anemias. At
controls were negative. The first account of what we the time of the discovery of the antiglobulin test, there
now call the indirect antiglobulin test was published by was great difficulty in distinguishing hemolytic
Coombs, Mourant, and Race in 1945.88 The authors anemia that was familial from that which was
were bold enough to state, “This test may have useful acquired. The only laboratory test available was the
applications in detecting fine degrees of sensitization in measurement of osmotic fragility, which was abnor-
other antigen-antibody systems. . . .” This has turned mal in familial hemolytic icterus (now called hereditary
out to be an understatement, for quite apart from the spherocytosis). However, Dameshek and Schwartz74
tests on red cells and bacteria covering all the isotypes pointed out that spherocytes causing increased
of antibody, an antiglobulin step or stage is a regular osmotic fragility could develop in cases that were
component in very many immunoassay procedures.85 clearly acquired hemolytic anemia.
A more substantial paper89 was published in the Barbara Dodd described the fact that she and
same year in the British Journal of Experimental Kathleen Boorman, who were working at the South
Pathology, and just as the printer’s page proofs were London Transfusion Centre with the director, John
on the point of dispatch back to the publisher, Loutit, who was already an authority in the field of
Mourant came across a paper in the German literature anemias, were in a privileged position.95 They had
from 1908 by Carlo Moreschi90 (Fig. 1-28) that visited Cambridge, where Race revealed to them the
described enhancement of red cell agglutination with secrets of the antiglobulin test before it had appeared in
an “antiserum to serum.” An acknowledgment was print. Dodd states that, “I shall never forget the gleam
22 Immune Hemolytic Anemias

FIGURE 1-25. (A) Patrick Mollison. (B) A 1947 photograph taken at the
Lister Institute in London showing, from left to right: Louis K. Diamond
whose research clarified the pathogenesis of hemolytic disease of the
fetus and newborn as well as the optimal management of that disorder;
Patrick L. Mollison, a pioneer in the field of blood transfusion and editor of
ten editions of the famous text, Blood Transfusion in Clinical Medicine;
Robert R. Race, an eminent immunohematologist who, along with his
long-time collaborator, Ruth Sanger, made innumerable contributions to
the field of RBC genetics and serology; and Sir Ronald A. Fisher, a famous
geneticist/biostatistician who, together with Race, devised a classification
of the Rh blood group system that is still used. (Courtesy of Professor
P. L. Mollison.)
A

100

FIGURE 1-26. Dacie and Mollison, using the Ashby technique,


were the first to demonstrate that normal RBCs survive nor- 80
mally in patients with familial hemolytic anemia. The figure
Percentage survival

shows survival of RBCs from normal donors after transfusion


to six patients with familial hemolytic anemia. Case 3 was an
Rh-negative patient who was later found to have developed an 60
5
alloantibody to Rh, accounting for the shortened survival of 2
transfused Rh-positive RBC. Although not shown in the figure,
survival in cases 2 and 5 was followed to completion and
found to exceed 100 days in each case. The dotted lines indi- 40 Cas
e1
cate the limits of survival in a group of normal recipients
(Mollison, unpublished observations). (From Dacie JV, Ca
Mollison PL: Survival of normal erythrocytes after transfusion se
to patients with familial haemolytic anaemia (acholuric jaun- 20 4
Ca

dice). The Lancet, volume i, May 1, 1943, pp 550–552.)


se
3

Case
6
0
0 20 40 60 80 100 120
Days after transfusion
Historical Concepts of Immune Hemolytic Anemias 23

FIGURE 1-27. Robin R. A. Coombs. (Photograph by Lawrence E. Young


M.D., Fellows’ Garden, Kings College, Cambridge University, 1950.
From Packman CH: The spherocytic haemolytic anaemias. Br J FIGURE 1-28. A photographic portrait of Carlo Moreschi. (From
Haematol 2001;112:888–899.) Coombs RR: Historical note: Past, present and future of the anti-
globulin test. Vox Sang 1998;74:67–73.)

in his eye when we returned from Cambridge with a interest in the agglutination or in Moreschi himself.
description of the new test!” They quickly collected the However, 6 months after the lecture was published in
RBCs of 17 patients with familial hemolytic anemia the Italian medical journal l’Informatore Medico,97
and 5 others with hemolytic anemia of the acquired Dr. Coombs received a letter from Dr. Pietro de
type. “It was enormously exciting then, but no surprise Ruggieri, who was a steroid chemist in Milan and who
now, to find that the 5 patients having acquired type was a nephew of Carlo Moreschi. He was delighted
had positive DATs, whereas the 17 familials were neg- with the reference to his long-since-dead uncle.
ative.” They concluded (correctly) that the agglutina-
tion tests “will discriminate the congenital from the
acquired form [of hemolytic icterus], and that it indi-
THE CONCEPT OF AUTOIMMUNE
cates that the acquired form is due to a process of HEMOLYTIC ANEMIA
immunization, whereas the congenital form is not.”
Thus, not only had they found a test that would distin- In 1951, Young and associates98 were the first to coin
guish between the familial and acquired forms of the term autoimmune hemolytic anemia. It was theorized
hemolytic anemia, but they had also demonstrated a that the production of an autoantibody was the result
difference in their etiology. of a breakdown in the “regulatory contrivances,” thus
A Note about Carlo Moreschi. Carlo Moreschi was leading to autoimmunization. However, the concept
deep in immunological research at Pavia at the turn of that a patient could produce autoantibodies was
the twentieth century. He published two particularly vigorously resisted by some. Witebsky,99 in particular,
interesting papers90,96 describing enhancement of was reluctant to draw the conclusion that the RBC
agglutination with antiserum to serum (i.e., with anti- coating material demonstrated by the antiglobuin test
globulin) (Table 1-1). However, incomplete antibodies was a true autoantibody. He considered it unproved
were unknown at the time and general acceptance or that the RBC could be involved in autoimmunization,
use of this procedure never resulted. Dr. Coombs paid with the implied breaking of the principle of horror
tribute to Moreschi and his researches in a lecture to the autotoxicus. This reluctance to accept the autoim-
Italian Association of Medical Analysts and Patholo- mune nature of antiglobulin test−positive hemolytic
gists entitled “Moreschi and Some Recent Develop- anemias led to the use for a time of the noncommittal
ments in Agglutination.” There seemed to be little term “antiglobulin-positive hemolytic anemia.”100
24 Immune Hemolytic Anemias

TABLE 1-1. TRANSLATED FROM MORESCHI (1908), DEMONSTRATING THE PRINCIPLE


OF THE ANTI-GLOBULIN (COOMBS) REACTION

Agglutination with
Goat Immune Serum or Rabbit Precipitating
Rabbit RBCs Goat Normal Serum Serum Immune Serum Normal Serum

1 mL 0.005 mL 0.0001 mL 0 0
1 mL 0.005 mL 0.005 mL Scant 0
1 mL 0.005 mL 0.001 mL Marked 0
1 mL 0.005 mL 0.005 mL Very marked 0
1 mL 0.005 mL 0.01 mL Very marked 0
1 mL 0.005 mL 0.05 mL Very marked 0
1 mL 0.005 mL 0.1 mL Very marked 0
1 mL – 0.1 mL 0 0
1 mL 0.01 mL – 0 0
2 hr room temperature Cells centrifuged and washed with normal saline 2 hr room temperature

Rabbit RBCs were incubated with goat immune serum, washed, and incubated with rabbit antibody to goat serum (precipitating serum). The RBCs agglutinated in a
dose-dependent manner. The controls, lacking either goat immune serum or rabbit precipitating serum, showed no agglutination.
Reproduced with modification from Packman CH: The spherocytic haemolytic anaemias. British Journal of Haematology 112:888–899.

Through the extensive writings and teaching of is not an ideal label because of the elution of the label
such eminent physicians as Dameshek, the concept of from the RBCs. The nearest rival to 51Cr is DF32P,
an autoimmune etiology for some types of acquired which was first reported in 1954 to be a potentially a
hemolytic anemias gradually obtained general recog- satisfactory label for RBCs.104 The DF32P technique has
nition and application.1 the advantage over 51Cr in that once attached to the
RBCs, it is not eluted.
The elimination curve of normal RBCs in a healthy
RADIOACTIVE CHROMIUM recipient, as demonstrated by the Ashby method or by
(51CR) AND DF32P the use of DF32P, is virtually a straight line, and this is
consistent with the concept of gradually increasing
The first studies using 51Cr were reported by Gray and senescence rather than of random elimination in which
Sterling101 in 1950 from Boston. They found that the the cells would be destroyed indiscriminately regard-
labeled RBCs lost radioactivity at a rate more rapid less of age. Indeed, the analysis of survival curves has
than could be predicted from the known normal life contributed most significantly to the understanding of
span of dog RBCs and, consequently, did not recognize the pathogenesis of increased hemolysis.3
the potential usefulness of the method in determining
long-term RBC survival.102 Later, Ebaugh and cowork- COLD AGGLUTININ SYNDROME (CAS)
ers103 labeled normal blood with 51Cr and transfused it
into normal human volunteers. Subsequently, the Cold agglutinins were initially demonstrated by
amount of radioactivity per milliliter of RBCs was Landsteiner in animal blood in 1903105 and in human
quantitated and a simultaneous evaluation was made blood by Mino in 1924,106 but their significance in
of the RBC survival by the Ashby differential aggluti- human disease was not accurately appreciated until
nation technique. They found that the two curves several decades later. The first determination of titers
reached extinction point at the same time. Calculations in an acute postpneumonic cold agglutinin disease
of the two curves were consistent with the hypothesis was made by Clough and Richter in 1918.107 A recog-
that chromium was leaking from the RBCs in an expo- nition of the relationship between cold agglutinins,
nential fashion with a mean half-life of 77 ± 12 days. hemolytic anemia, Raynaud’s phenomenon, and
Correcting for this leakage, the curve for the two tech- hemoglobinuria began to emerge with the case reports
niques approximated that determined by the straight- of Iwai and Mei-Sai in 1925 and 1926.108,109 Their first
line Ashby differential agglutination survival curve.103 patient was a 36-year-old Chinese man giving a 6-year
The value of the isotope as a harmless label of RBCs
was soon confirmed in many centers throughout the *As mentioned in Chapter 2, describing the skin manifestations in
world, and because the 51Cr could be used to label cold agglutinin syndrome as Raynaud’s phenomena is, strictly
patients’ own RBCs and to study their survival in speaking, incorrect.110 Raynaud’s disease, the consequence of
their own circulation, as well as to label transfused vasoconstriction, leads in sequence of three phenomena: First, the
blood, Ashby’s elegant but laborious technique, with affected part becomes white and perhaps numb; then it becomes
its inherent limitations and technical difficulties soon swollen, stiff and livid; and finally, when the vasoconstriction
passes off, the part becomes red due to reactive hyperemia. In
became obsolete. CAS the changes, which preferably are termed acrocyanosis, or
51Cr is still widely used in studies of RBC life span
literally “blue extremity,” differ from those of Raynaud’s disease
and in the measurement or blood volume, although it in the absence of an initial white phase because there is no
Historical Concepts of Immune Hemolytic Anemias 25

history of Raynaud’s disease.* His serum contained a higher in vitro following the loss of CO2, and the anti-
cold agglutinin that reacted to a titer of 1,000 at 0°C bodies do not cause optimal lysis at alkaline pH.
and reacted up to 30°C against normal RBCs as well as Dacie118 demonstrated the presence of cold hemolysins
those of the patient. They demonstrated that the cir- in sera containing cold agglutinins by adding a trace of
culation of the patient’s blood through fine tubes was hydrochloric acid to produce a slightly acid pH value.
impeded when the blood was cooled to 5°C and sug- However, he still used the two-step temperature
gested that the Raynaud’s phenomenon might be re- arrangement in the classic Donath-Landsteiner test. In
lated to mechanical obstruction by autoagglutinated 1953 Schubothe pointed out that hemolysis caused by
RBCs. In their second patient, a woman aged 78, they the cold agglutinins does not have a bithermic mode of
showed that cooling of the fingers was associated action but takes place monothermically.119,120 He intro-
with breaking of the column of blood in the capillaries duced the term cold hemagglutinin disease to separate the
of the nail bed. However, in neither case did the disorder from other acquired hemolytic anemias.
authors describe hemoglobinuria or anemia. In the 1950s it ultimately became apparent that there
Druitt,113 writing from Madras in 1873, described in existed an obscure and rather unusual syndrome,
detail the history of a doctor, aged 51 years, who over a which affected almost exclusively elderly subjects, that
period of at least 6 years had experienced attacks of was characterized by mild to moderate hemolytic
numbness of the feet and a purplish blue discoloration anemia and by the presence in the patient’s serum of
of the hands on exposure to cold. These attacks might be cold agglutinins at high titers, so that massive and
followed by the passage of “hematinuria.” The patient rapid autoagglutination took place if their blood, after
obtained relief from his symptoms when he went to live withdrawal, was allowed to cool to room temperature.
in a warm climate (India). Druitt believed that the In cold weather the patients suffered from what was
nervous system and the blood were involved and sug- often described as Raynaud’s phenomenon, affecting
gested that the blood was undergoing “a hemolysis, a the fingers, toes, and earlobes, and sometimes this led
decomposition or necrosis of the blood globules.” to local gangrene. Hemoglobinuria, too, often devel-
Roth, in 1935, reported a 59-year-old man who oped in cold weather. This is the condition we now
suffered from Raynaud’s phenomenon affecting his refer to as cold CAS.
hands, feet, and nose when exposed to mild degrees Discovery of Blood Group Specificity of Pathologic
of cold.114 More severe chilling produced hemoglobin- Cold Agglutinins. Early studies of the specificity of the
uria. The author noted that the patient’s blood under- cold agglutinin in patients with CAS demonstrated no
went rapid autoagglutination after withdrawal, which blood group specificity. Mino121 is usually quoted as
was reversed by warming. having introduced the concept of the “nonspecific”
In the same year, Ernstene and Gardner115 reported nature of cold agglutinins; he concluded that all human
a 38-year-old man who had attacks of hemoglobinuria RBCs shared a common receptor and that no distinc-
and Raynaud’s phenomenon on exposure to cold. tion could be made with regard to reactivity between
Autoagglutination of his blood was noted at room cells of different ABO groups. However, Wiener and his
temperature, red blood cell counts were difficult to coworkers122 reported in 1956 that they had tested a
perform, the cold agglutinin titer was 1280, and he serum derived from a patient with CAS against 22,964
was anemic with a hemoglobin of 10.5 g/dL. blood samples! Five samples only, as well as the
Despite these early reports, CAS did not receive patient’s own RBCs, were not agglutinated at room
wide recognition and the pathogenetic role of cold temperature. The insensitive RBCs were designated “i”
agglutinins was not well accepted. Indeed, as late as or “I-negative,” and the serum was said to contain
1943, Stats and Wasserman116 published a review in “anti-I” (“I” for individuality). Thus started the unrav-
which they stated that in the great majority of cases eling of the complex Ii blood group system (see
cold hemagglutination was innocuous, although “in Chapter 6). By far the most common type of high-titer
some cases” of hemolytic anemia, PCH, Raynaud’s cold antibody reacts with the I antigen, a small minor-
syndrome, and peripheral gangrene, the cold hemag- ity with the i antigen, and a few antibodies react with
glutination is of pathogenetic significance. Accurate antigens other than I and i (see Chapter 7).
descriptions of the syndrome and features that distin- The Physical Nature of Cold Agglutinins. The anti-
guished it from other forms of AIHA appeared during bodies also have been studied by physical means. First,
the 1950s.117 the use of the ultracentrifuge showed that in sera
The hemolytic activity of serum of patients with cold containing large amounts of a cold autoantibody, this
agglutinin disease had not been well recognized would separate as a high-density protein and might
because the pH of blood rapidly rises to pH 8 and also be visualized as a distinct sharp peak in the beta-
gamma region on simple paper electrophoresis.123
vasoconstriction, and in that the blue cyanotic phase is more Subsequently, when methods of immunoelectro-
intense; the affected part may in fact become deep purple. There phoresis became available, it was clearly shown that not
is, too, no final hyperemic phase. Marshal et al.111 and Hillestad112 only were these protein peaks composed of macro-
showed that the blood flow reactions to chilling are quite distinct
from those in Raynaud’s disease proper. No evidence of an globulin (IgM) but that they were also monoclonal. In
abnormal vascular response could be obtained. Both processes that respect CAS is analogous to Waldenström’s
can, however, lead to local gangrene. macroglobulinemia in that the basis of both disorders is
26 Immune Hemolytic Anemias

the formation by the patient of large amounts of an IgM There is an apocryphal story that when Pope
paraprotein. Innocent VIII was on his deathbed in 1492, a last des-
Subsequently, numerous case reports and detailed perate attempt at his survival was made on the recom-
reviews of clinical findings, laboratory features, sero- mendation of an unknown physician. He received the
logic and immunochemical characterization of the blood of three youths supposedly via transfusion,
antibodies, and the pathogenesis of CAS have been although more likely as a draught. The fact is that
published (see Chapter 3). shortly thereafter he passed on, to Heaven, doubtlessly.
The prescribing physician wisely and quickly disap-
peared⎯in which direction is not recorded.125
MORE RECENT EVENTS Early Suggestions for Transfusions. Up to the seven-
teenth century, blood must have been given only by
The investigators who, in the early days, contributed to mouth. Direct transfusion into the circulation had to
our understanding of AIHA as we know it today were await the discovery that there was a circulation. The
clinicians in the true sense of the word. They studied at beginnings of transfusion therapy date from the mid-
the bedside and in clinical laboratories, using their seventeenth century following Harvey’s momentous
minds, hands, eyes, and ears; their most sophisticated discovery of the circulation of the blood. He announced
instrument was a microscope. Information transmittal in a monumental treatise, De Motu Cordis, that blood
and retrieval were rudimentary at best; if the journals circulated within the body in a closed system, main-
were available, the language was more probably tained by the heart acting as a pump, and that the blood
foreign to the reader than not, either French, German, was sent to the limbs through the arteries and returned
or English. They made errors, but they also identified through the veins, whose valves did not oppose its
and corrected them, so as to lay a foundation for the course that way. This stimulated actual experimenta-
more sophisticated studies that were to come. tion with injections into the bloodstream.125
The second half of the twentieth century brought
important new insights into the diagnosis, pathogene-
sis, and management of AIHA. Important advances FIRST RECORD OF TRANSFUSIONS
occurred concerning the roles of RBC structure and bio-
chemistry, the specificity of autoantibodies and their The first well-documented transfusions were carried
molecular structure, the molecular nature and reaction out by two widely separated investigators, one
mechanism of serum complement, the concept of drug- English, the other French. Because both individual
induced immune hemolytic anemias including drug- and national priorities were at stake, considerable
induced AIHA, mechanisms of hemolysis, RBC controversy was engendered and numerous publica-
structure, and its genetic regulation. Future years will tions resulted as to who should be accredited with
undoubtedly bring new understandings of pathogene- doing the first transfusion.128-131
sis at the molecular and genetic level, and new means In England, a young physiologist and physician,
of treatment, possibly involving the sciences of stem Richard Lower (Fig. 1-29), of Oxford, participated in
cell transplantation and gene replacement therapy. experiments of injecting opiates, emetics, and other
medicines into the veins of living animals. As he stated
in letters then and in a book published later, this stimu-
HISTORICAL NOTES REGARDING lated ideas about injecting large quantities of blood
HEMOLYTIC TRANSFUSION REACTIONS from different animals. In February 1665, he developed
the needed surgical skill and performed his first suc-
The fascinating history of blood transfusion has been cessful transfusion, from the cervical artery of one dog
reviewed in a number of publications124-127 and, into the jugular vein of another, previously almost ago-
among descriptions of the early attempts at transfu- nally exsanguinated. The recipient animal was
sion therapy, are dramatic accounts of hemolytic promptly restored to a healthy active state. There was
transfusion reactions. This is to be expected because no untoward reaction, for dogs do not have natural
transfusions were carried out long before there was isoagglutinins, although they do vary in blood group
knowledge of blood groups or current good manufac- antigens. Lower’s experiments were recorded in the
turing practices. Journal des Savants of January 31, 1667.124
The Early History of Blood as a Therapeutic
Measure. Blood, in one form or another, was men- THE FIRST RECORDS OF HEMOLYTIC
tioned as a possible therapeutic measure throughout
ancient times. The Egyptians were said to advocate TRANSFUSION REACTIONS
blood baths for purposes of recuperation and rejuve-
nation. As late as the fifteenth century, blood was rec- In France, a philosopher-mathematician and physician,
ommended to remedy a variety of ailments, such as Jean-Baptiste Denis (Fig. 1-30), performed the first
lunacy, fits, palsy, melancholia, and bad disposition, transfusion of a human on June 15, 1667. His patient
but not for blood loss or anemia, as would have was a boy of 15, a sufferer from a prolonged febrile
seemed more logical. illness and profound lethargy. He had been subjected
Historical Concepts of Immune Hemolytic Anemias 27

FIGURE 1-29. Richard Lower


(1631–1691). Oil painting by Jacob
Huysmans. (From Moore P: Blood and
Justice. Chichester, England: John Wiley &
Sons Ltd., 2003.)

to, and had somehow managed to survive, 20 phle- later the procedure was repeated. This time, there
botomies. Denis succeeded in transfusing him with developed all the signs now recognized as typical of a
about 9 ounces of sheep’s blood and actually “cured” severe hemolytic transfusion reaction. Denis’s descrip-
him of his ailment. Encouraged by this success, Denis tion can be considered a medical classic132:
tried his good fortune again. This time he used a
healthy paid volunteer who received 20 ounces of As soon as the blood began to enter into his veins, he
sheep’s blood without recorded difficulties except for felt the heat along his arm and under his armpits. His
feeling “very great heat” along the vein in his arm and pulse rose and soon after we observed a plentiful sweat
later voiding “black urine.” Although the black urine over all his face. His pulse varied extremely at this
instant and he complained of great pains in his kidneys,
strongly suggests a hemolytic transfusion reaction, he and that he was not well in his stomach, and that he
was otherwise asymptomatic and was so little dis- was ready to choke unless given his liberty. He was
turbed that he proceeded to butcher the sheep and then made to lie down and fell asleep, and slept all night
went off on a drinking bout with companions.125 without awakening until morning. When he awakened
A third subject, a Swedish nobleman already mori- he made a great glass full of urine, of a color as black as
bund, did not fare so well and died soon after an if it had been mixed with the soot of chimneys.124
attempted transfusion.
Next Denis treated a man who had episodes of Denis recounted that the following morning the
violent maniacal behavior. The transfusion was on subject also manifested hemoglobinuria and had epis-
December 19, 1667, with 5 or 6 ounces of blood from the taxis. However, by the third day his urine cleared, and
femoral artery of a “gentle calf,” which “might dampen he improved his mental status and returned to his
his spirits.” The patient seemed to improve. A few days wife. Denis attributed the color of the urine to a “black
28 Immune Hemolytic Anemias

via silver tubes and quills to a sheep’s carotid artery. It


was surmised that during 2 minutes, 9 to 10 ounces of
blood were so transferred. The patient afterward
“found himself very well” and 6 days later gave the
society a talk in Latin telling how much better he felt.
Nowhere was any comment recorded about the effect
of the transfusion on the patient’s temperament or his
“too warm brain.”125

NATIONAL AND INTERNATIONAL


CONTROVERSY

In an action that presages modern medicine, the wife


of the patient who was transfused by Denis sued
him, charging that the transfusion had killed her
husband.124,125,127 Considerable furor was raised
among Parisian physicians, but at the trial the
defense was successful in proving that the man had
been poisoned with arsenic by his wife. Although
Denis was thus exonerated, the Paris Society of
Physicians declared itself against such experiments
and persuaded the criminal court in Paris on April
17, 1668, to forbid further transfusions without
approval from the Faculty of Medicine of Paris,
known to be bitterly opposed to the procedure. Ten
years later, an edict of Parliament prohibited transfu-
sion experiments on humans. Soon thereafter, the
Royal Society in England disapproved transfusion
practices, as did the magistrates in Rome. This
eclipse of overt interest in transfusion therapy lasted
150 years.125
In the meantime, an international debate had been
initiated as to who and which country should be cred-
ited with the first transfusion. Throughout 1667 and
1668, many around Europe contributed to the debate in
the form of letters and published pamphlets. Most fell
neatly into pro-Denis or anti-Denis camps, although a
few were prepared to express an open mind. The con-
FIGURE 1-30. Jean-Baptiste Denis (From Moore P; Blood and Justice. troversy is reviewed in detail by Moore.127
Chichester, England: John Wiley & Sons Ltd., 2003.) England’s claim was based on Lower’s thoroughly
documented dog-to-dog transfusions in 1665. The
French claimed that the idea had been proposed
choler” that had been retained in the body and had 10 years earlier and that human transfusions were
sent vapors to the brain that caused the subject’s first done by Denis. National prestige seemed to be at
mental disturbance.132 Several months later the stake even though the treatment was admittedly less
patient again became violent and irrational and his than uniformly successful. A considerable exchange
wife insisted on yet another transfusion. Denis of letters between Denis and Henry Oldenburg,133
attempted this but without success because the man the secretary of the Royal Society, took place in late
was violent and would not cooperate. He died the fol- 1667 and 1668 with publication in the Proceedings of
lowing night. the Society. Denis had sent a letter to the Philosophical
By this time, Lower had also initiated transfusion in Transactions, in London, the official publication of the
humans. On November 23, 1667, he and his skilled Royal Society, describing his first transfusion and
associate Edmund King performed their first human this was actually printed dated July 22, 1667.
transfusion before The Royal Society. The patient was a However, its publication did not take place until
22-year-old member of the clergy who was “somewhat September because the editor, Oldenberg, was incar-
unbalanced, whose brain was considered a little too cerated in the Tower of London on suspicion of
warm.” It was hoped that the operation would alter his treason. Fortunately, he was declared innocent. (Few
character. Accordingly, he was bled from his antecu- editors can claim so valid an excuse for delays in
bital vein for 6 or 7 ounces and then he was connected publication.125)
Historical Concepts of Immune Hemolytic Anemias 29

Nevertheless, considering that Lower did not 16. Creite A: Versuche uber die Wirkung des Serumeiweisses nach
perform his first human transfusion until November of Injection in das Blut. Zeitschrift für Rationelle Medicin
1869;36:90–108.
that year, there seems little question that Denis was 17. Landois L: Die Transfusion des Blutes. Leipzig, 1875.
the first to perform transfusion of a human being.124 17a. Gottlieb AM, Karl Landsteiner: The melancholy genius: His
The best that Oldenburg could contend was that the time and his colleagues, 1868–1943. Transfus Med Rev
“English might well have been first if they had not been 1998;12:18–27.
17b. Schwarz HP, Dorner F: Karl Landsteiner and his major contri-
so tender in hazarding the life of man, “a post hoc solic- butions to haematology. Br J Haematol 2003;121:556–565.
itude with no foundation in fact.125 The controversy 18. Landsteiner K: Zur Kenntness der antifermentiven lytischen
regarding priority long remained in doubt and was not und agglutinierenden Wirkungen des Blutserums und der
really resolved satisfactorily. It finally seemed to be Lymph. Centralblatt für Bacteriologie 1900;27:357–366.
accepted that Lower, of England, deserved the credit 19. Landsteiner K: Uber Agglutinationserscheinungen normal
menschlichen Blutes. Wiener Kinische Wochenschrift
for doing and fully describing the first animal transfu- 1901;14:1132–1134.
sions, whereas Denis, of France, was credited with the 20. Diamond LK: The story of our blood groups. In Wintrobe MM
first successful transfusions in humans.125 Denis should (ed): Blood, Pure and Eloquent. New York: McGraw-Hill,
also be credited with the first accurate and detailed 1980:691–717.
21. Tagarelli A, Piro A, Lagonia P, Tagarelli G: Karl Landsteiner: A
description of a hemolytic transfusion reaction! hundred years later. Transplantation 2001;72:3–7.
It was not until the late 19th century that successful 22. Garratty G: Immunohematology is 100 years old. J Lab Clin
transfusions were reported, again by an English obste- Med 2000;135:110–111.
trician, William Blundell. Transfusion did not become 23. Vanlair CF, Masius JR: De la microcythemie. Bull Acad R Med
commonly used until almost a decade following Belg 3e Ser 1871;5:515–613.
24. Hayem G: Sur une variete particuliere d’ictere chronique.
Landsteiner’s discovery of the ABO blood groups.124–126 Ictere infectieux chronique splenomegalique. Presse Med
1898;6:121–125.
25. Minkowski O: Ueber eine hereditare, unter dem bilde eines
chronischen icterus mit urobilinurie, splenomegalie und
R E F E R E N C E S nierensiderosis verlaufende affection. Verhandl Krong f Inn
Med 1900;18:316–321.
1. Pirofsky B: The hemolytic anemias—historical review and 26. Crosby WH: The pathogenesis of spherocytes and leptocytes
classification. In Pirofsky B (ed): Autoimmunization and the (target cells). Blood 1952;7:261–274.
Autoimmune Hemolytic Anemias. Baltimore, MD: Williams 27. Chauffard A: Pathogenie de l’ictere congenital de l’adulte.
& Wilkins, 1969:3–20. Semaine Med 1907;27:25–29.
2. Dacie JV: Auto-immune haemolytic anaemia AIHA: Warm- 28. Haden RL: The mechanism of the increased fragility of the
antibody syndromes. I: “idiopathic” types: History and clini- erythrocytes in congenital hemolytic jaundice. Am J Med Sci
cal features. In The Haemolytic Anaemias, 3rd ed., vol. 3, The 1934;188:441–449.
Auto-Immune Haemolytic Anaemias. New York: Churchill 29. Chauffard A, Fiessinger N: Ictere congenital hemolytique avec
Livingstone, 1992:6–53. lesions globulaires. Soc Med Hosp Paris 1907;24:1169–1178.
3. Dacie JV: The life span of the red blood cell and circum- 30. Chauffard A: Les icteres hemolytique. Semaine Medicale
stances of its premature death. In Wintrobe MM (ed): Blood, 1908;28:49–52.
Pure and Eloquent. New York: McGraw-Hill, 1980:211–255. 31. Lajtha LG: The common ancestral cell. In Wintrobe MM (ed):
4. MacK P, Freedman J: Autoimmune hemolytic anemia: A Blood, Pure and Eloquent. New York: McGraw-Hill, 1980:81–95.
history. Transfus Med Rev 2000;14:223–233. 32. Vaughan VC: On the appearance of certain granules in the
5. Dacie SJ: The immune haemolytic anaemias: A century of erythrocytes of man. J Med Res 1903;10:342–366.
exciting progress in understanding. Br J Haematol 2001; 33. Ehrlich P, Morgenroth J: Six landmark communications on
114:770–785. hemolysis. Berl Klin Wochenschr 1899;36:6 and 481; 1900;
6. Nydegger UE, Kazatchkine MD, Miescher PA: Immunopatho- 37:453 and 681; 1901;38:251 and 569. (These also appear in The
logic and clinical features of hemolytic anemia due to cold Collected Papers of Paul Ehrlich, vol 2. New York: Pergamon,
agglutinins. Semin Hematol 1991;28:66–77. 1957, in both German and English translation, and in English
7. Packman CH: The spherocytic haemolytic anaemias. Br J alone in Collected Studies on Immunity, C. Bolduan, transla-
Haematol 2001;112:888–899. tor, New York: Wiley, 1906.)
8. Silverstein AM: The Donath-Landsteiner autoantibody: The 34. Bordet J: Ann Inst Pasteur, Paris 1898;12:688.
incommensurable languages of early immunologic dispute. 35. Ehrlich P: The Collected Papers of Paul Ehrlich, vol. 2,
Cell Immunol 1986;97:173–188. pp. 298–315. New York: Pergamon, 1957.
9. Wintrobe MM: Blood, Pure and Eloquent. New York: 35a. Silverstein AM: Autoimmunity versus horror autotoxicus:
McGraw-Hill, 1980. The struggle for recognition. Nat Immunol. 2001;2:279–281.
10. Wintrobe MM: The lessons of history. In Wintrobe MM (ed): 36. Ehrlich P: The Collected Papers of Paul Ehrlich, vol. 1,
Blood, Pure and Eloquent. New York: McGraw-Hill, p. 253. New York: Pergamon, 1957.
1980:719–726. 37. Dacie JV: Auto-immune haemolytic anaemia AIHA: Cold-
11. Galen C: Volume VIII. In Kuhn DCG (ed): Opera Omnia. antibody syndromes. V: paroxysmal cold haemoglobinuria
Lipsiae 1824:356. (PCH). In The Haemolytic Anaemias, 3rd ed., vol. 3, The
12. Wintrobe MM: Milestones on the path of progress. In Auto-Immune Haemolytic Anaemias. New York: Churchill
Wintrobe MM (ed): Blood, Pure and Eloquent. New York: Livingstone, 1992:329–362.
McGraw-Hill, 1980:1–31. 38. Heddle NM: Acute paroxysmal cold hemoglobinuria.
13. Andral G: Essai d’Hematologie Pathologique. Paris, France: Transfus Med Rev 1989;3:219–229.
Fortin et Masson, 1943. 39. Rosenbach O: Zur Leher von der periodischen Hamoglobin-
14. Vogel J: Upon the colour of the urine. Med Tmes Gazette urie. Dtsch Med Wschr 1879;5:613.
1853;7:378. 40. Rosenbach O: Beitrag zur Lehre von der periodischen
15. Hughes-Jones NC, Gardner B: Red cell agglutination: The first Hamoglobinurie. Berl Klin Wschr 1880;17:132, 151–153.
description by Creite (1869) and further observations made by 41. Dressler: A case of intermittent albuminuria and chromaturia.
Landois (1875) and Landsteiner (1901). Br J Haematol In Major RH (ed): Classic Descriptions of Disease. Springfield,
2002;119:889–893. IL: Charles C Thomas, 1939:590–592.
30 Immune Hemolytic Anemias

42. Elliotson J: Diseases of the heart united with ague. Lancet 74a. Dacie JV: The Haemolytic Anaemias, 3rd ed. vol. 3, The
1832;i:500–501. Auto-Immune Haemolytic Anaemias. New York: Churchill
43. Wiltshire A: Urine from a case of intermittent haematuria. Livingstone, 1992.
Trans Pathol Soc Lond 1867;18:180. 75. Rous P: Destruction of the red blood corpuscles in health and
44. Secchi: Ein Fall von Hamoglobinurie aus der Klinik des Geh. disease. Physiol Rev 1923;3:75–105.
Rath Prof. Dr. Lebert. Berl Klin Wochenschr 1872;9:237–239. 76. Ashby W: The determination of the length of life of transfused
45. Mackenzie S: Paroxysmal haemoglobinuria, with remarks on blood corpuscles in man. J Exp Med 1919;29:267–281.
its nature. Lancet 1879;2:116, 155–117, 157. 77. Ashby W: Some data on the range of life of transfused blood-
46. Kuessner B: Paroxysmale hamaglobinurie. Dtsch Med corpuscles in persons without idiopathic blood diseases. Med
Wochenschr 1879;5:475–478. Clin North Am 1919;3:783–799.
47. Mackenzie S: On paroxysmal haemaglobinuria. Lancet 78. Ashby W: Study of transfused blood. I. The periodicity in
1884;1:156, 198, 243–158, 200, 245. eliminative activity shown by the organism. J Exp Med
48. Ehrlich P: Uber paroxymale Hamoglobinurie. Dtsch Med 1921;34:127–146.
Wschr 1891;7:224–225. 79. Landsteiner K, Levine P, Janes ML: On the development of
49. Donath J, Landsteiner K: Uber paroxysmale hamoglobinurie. isoagglutinins following transfusion. Proc Soc Exp Biol Med
Munchen Med Wochenschr 1904;51:1590-1595. 1928;25:672–674.
50. Bibel DJ: On paroxysmal hemoglobinuria. 1904, Julius Donath 80. Wiener AS: Longevity of the erythrocyte. JAMA 1934;
and Karl Landsteiner. In: Bibel DJ (ed): Milestones in 102:1779–1780.
Immunology. Berlin: Springer-Verlag, 1988:63–67. 81. Dedichen HG: Ictere hemolytique et ulcere de la jambe. Acta
51. Bird GW, Wingham J, Martin AJ, et al: Idiopathic non-syphilitic Med Scand 2003;77:411–430.
paroxysmal cold haemoglobinuria in children. J Clin Pathol 81a. Vaughan J: John Dacie: Br J Haematol 1972;23(Suppl), p 7.
1976;29:215–218. 82. Dacie JV, Mollison PL: Survival of normal erythrocytes after
52. Eason J: The pathology of paroxysmal haemoglobinuria transfusion to patients with familial haemolytic anaemia
preliminary communication. Edin Med J 1906;19:43–52. acholuric jaundice. Lancet 1943;1:550–552.
53. Eason J: The pathology of paroxysmal haemoglobinuria. 83. Loutit JF, Mollison PL: Haemolytic icterus (acholuric jaundice)
J Pathol Bact 1906;11:167–202. congenital and acquired. J Path Bact 1946;58:711.
54. Donath J, Landsteiner K: Z Klin Med 1906;58:173. 84. Coombs RR: Historical note: past, present and future of the
55. Goltz D: Das Donath-Landsteiner Hamolysin. Die Entstehung antiglobulin test. Vox Sang 1998;74:67–73.
eines Mythos in der Medizin des 20 Jahrhunderts. Clio Med 85. Coombs RR: Immunohaematology: reminiscences and reflec-
1982;16:193. tions. Transfus Med 1994;4:185–193.
56. Rossle R: Ergeb Allg Pathol 1909;13:124, 228. 86. Race RR: An “incomplete” antibody in human serum. Nature
57. Meyer E, Emmerich E: Dtsch Arch Klin Med 1909;96:287. 1944;153:771–772.
58. Donath J, Landsteiner K: Uber paroxysmale Haemoglobin- 87. Weiner AS: A new test blocking test for Rh sensitization. Proc
urie. Munchen Med Wschr 1904;51:1590. Soc Exp Biol NY 1944;56:173–176.
59. Chauffard A, Troissier J: Semaine Med 1908;28:345. 88. Coombs RRA, Mourant AE, Race RR: Detection of weak and
60. Chauffard A, Vincent C: Hemoglobinurie hemolysinique avec “incomplete” Rh agglutinins: A new test. Lancet 1945;2:15–16.
ictere polycholique aigu. Semaine Med 2003;29:601–604. 89. Coombs RRA, Mourant AE, Race RR: A new test for the detec-
61. Widal F, Abrami P, Brule M: Les icteres d’origine hemolytique. tion of weak and “incomplete” Rh agglutinins. Br J Exp Pathol
Arch Mal Coeur 1908;1:193–231. 1945;26:255–266.
62. Widal F, Abrami P, Brule M: Ictere hemolytique acquis, a 90. Moreschi C: Neue Tatsachen uber die Blutkorperchen-
rechutes: Origine intestinale du processus hemolitique. Bull Agglutination. Zentralbl Bakteriol Parasitenkd Infektkr
Mem Soc Hop de Paris 1912;33:480–484. Originale 1908;46:49–51.
63. Du Role des Hemolysines en Pathologia. Deuxieme 91. Coombs RRA, Mourant AE, Race RR: In vivo isosensitisation
Question du XII Session du Congres Francais de Medicine, of red cells in babies with haemolytic disease. Lancet
Lyon. 1911. 1946;1:264–266.
64. Micheli F: Unmittelbare effekte der splenektomie bei einem 92. Coombs RRA, Mourant AE: On certain properties of antisera
fall von erworbenem hamolytischen splenomegalischen prepared against human serum and its various protein frac-
ikterus typus Hayem-Widal spleno-hamolytischer ikterus. tions: their use in the detection of sensitisation of human red
Wiener Klinische Wochenschr 1911;24:1269–1274. cells with “incomplete” Rh antibody, and on the nature of this
65. Banti G: La esplanomegalia hemolitica. Semaine Med antibody. J Pathol Bact 1947;59:105–111.
1912;32:265–268. 93. Dacie JV: Differences in the behaviour of sensitized red cells to
66. Crosby WH: The spleen. In: Wintrobe MM (ed): Blood, Pure agglutination by antiglobulin sera. Lancet 1951;2:954–955.
and Eloquent. New York: McGraw-Hill Book Company, 94. Dacie JV, Crookston JH, Christenson WN: “Incomplete” cold
1980:97–138. antibodies: role of complement in sensitization to antiglobulin
67. Lederer M: A form of acute hemolytic anemia probably of serum by potentially haemolytic antibodies. Br J Haematol
infectious origin. Am J Med Sci 1925;170:500–510. 1957;3:77–87.
68. Lederer M: Three additional cases of acute hemolytic infec- 95. Dodd BE: First tests with anti-human globulin on the red cells
tious. anemia. Am J Med Sci 1930;179:228–236. of patients suffering from haemolytic anaemia. Vox Sang
69. Brill IC: Acute febrile anemia: a new disease? Arch Intern Med 1984;46:183–184.
1926;37:244–247. 96. Moreschi C: Beschleunigung und Verstarkung der Bakterien-
70. Tileston W: Hemolytic jaundice. Medicine 1922;1:355–388. agglutination durch Antieiweiss-sera. Zentralbl Bakteriol
71. Dacie JV: The Haemolytic Anaemias. Part II. Auto-Immune Parasitenkd Infektkr Originale 1908;46:456–460.
Haemolytic Anaemias, 2nd ed. London: J. & A Churchill Ltd, 97. Coombs RRA: Moreschi e alcuni recenti sviluppi nello studio
1962. della agglutinazioni. Informatore Med 1954;9:126–129.
72. Dameshek W, Schwartz SO: The presence of hemolysins in 98. Young LE, Miller G, Christian RM: Clinical and laboratory
acute hemolytic anemia; preliminary note. N Engl J Med observations on autoimmune hemolytic disease. Ann Intern
1938;218:75. Med 1951;35:507–517.
73. Dameshek W, Schwartz SO: Hemolysins as the cause of 99. Witebsky E: Historical roots of present concepts of immuno-
clinical and experimental hemolytic anemias. With particular pathology. In: Grabar P, Miescher (eds): Immunopathology,
reference to the nature of spherocytosis and increased First International Symposium. New York: Grune & Stratton,
fragility. Am J Med Sci 1938;196:769. 1959.
74. Dameshek W, Schwartz SO: Acute hemolytic anemia (acquired 100. Pirofsky B: A new diagnostic test for antiglobulin positive
hemolytic icterus, acute type). Medicine 1940;19:231–327. (autoimmune) haemolytic anaemia. Br J Haemat 1960;6:395.
Historical Concepts of Immune Hemolytic Anemias 31

101. Gray SJ, Sterling K: Tagging of red cells and plasma proteins 117. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Cold-
with radioactive chromium. J Clin Invest 1950;29:1604–1613. antibody syndromes. I: ‘Idiopathic types’:clinical presentation
102. Ebaugh FG, Jr, Ross JF: The radioactive sodium chromate and haematological and serological findings. In: The
method for erythrocyte survival. Vox Sang 1985;49:304–307. Haemolytic Anaemias, 3rd ed. vol. 3, The Auto-Immune
103. Ebaugh FG J, Emerson CP, Ross JF: The use of rdioactive Haemolytic Anaemias. New York: Churchill Livingstone,
chromium-51 as an erythrocyte tagging agent for the deter- 1992:210–239.
mination of red cell survival in vivo. J Clin Invest 118. Dacie JV: The presence of cold haemolysins in sera containing
1953;32:1260. cold haemagglutinins. J Pathol Bact 1950;62:241.
104. Cohen JA, Warringa MGPJ: The fate of P32-labelled diiso- 119. Schubothe H: The cold hemagglutinin disease. Semin
propylfluorophosphonate in the human body and its use as a Hematol 1966;3:27–47.
labelling agent in the study of the turnover of blood plasma 120. Schubothe H: Serologische Besonderheiten unspezifischer
and red cells. J Clin Invest 1954;33:459–467. Saurekaltehamolysine. Klin Wochenschr 1953;31:808.
105. Landsteiner K: Uber Beziehungen zwischen dem Blutserum 121. Mino P: La panemoagglutinina del sangue umano. Policlin
und den Korperzellen. Munchen Med Wschr 1903;50:1812. Sez Prat 1924;31:1355–1359.
106. Mino P: Einiges uber die Konstitutionslehre und serologische 122. Wiener AS, Unger LJ, Cohen L, Feldman J: Type-specific cold
Forschung. Dtsch med Wschr 1924;50:1533. auto-antibodies as a cause of acquired hemolytic anemia and
107. Clough MC, Richter JM: A study of an autoagglutin occurring hemolytic transfusion reactions: Biologic tests with bovine red
in a human serum. Bull J Hopkins Hosp 1918;29:86. cells. Ann Intern Med 1956;44:221–240.
108. Iwai S, Mei-Sai N: Etiology of Raynaud’s disease; a prelimi- 123. Christenson WN, Dacie JV: Serum proteins in acquired haemo-
nary report. Jap Med World 1925;5:119. lytic anaemia auto-antibody type. Br J Haematol 1957;3:153–164.
109. Iwai S, Mei-Sai N: Etiology of Raynaud’s disease. Jpn Med 124. Oberman HA: The history of transfusion medicine. In: Petz
World 1926;6:345. LD, Swisher SN, Kleinman S, Spence RK, Strauss RG (eds):
110. Rosse WF: Clinical Immunohematology: Basic Concepts and Clinical Practice of Transfusion Medicine, 3rd ed. New York:
Clinical Applications. Boston: Blackwell Scientific Publications, Churchill Livingstone, 1996:11–32.
1990. 125. Diamond LK: A history of blood transfusion. In: Wintrobe
111. Marshall RJ, Shepherd JT, Thompson ID: Vascular responses MM (ed): Blood, Pure and Eloquent. New York: McGraw-Hill
in patients with high serum titres of cold agglutinins. Clin Sci Book Company, 1980:659–688.
1953;12:255–264. 126. Starr D: Blood: An Epic History of Medicine and Commerce.
112. Hillestad LK: The peripheral circulation during exposure to New York: Alfred A. Knopf, 1998.
cold in normals and in patients with the syndrome of high- 127. Moore P: Blood and Justice. Chichester, England: John Wiley
titre cold haemagglutination. II. Vascular response to cold & Sons Ltd, 2003.
exposure in high-titre haemagglutination. Acta Med Scand 128. Hoff HE, Guilleman R: The first experiments on transfusion in
1959;164:211–218. France. J Hist Med 1963;18:103–124.
113. Druitt R: Two cases of intermittent haematinuria. Med Tmes 129. Hollingsworth MW: Blood transfusion by Richard Lower in
Gazette London. 1873;i:408, 461, 489-411,462, 490. 1665. Ann Med Hist 1928;10:213–225.
114. Roth G: Paroxysmal hemoglobinuria with vasomotor and 130. Hutchin P: History of blood transfusion:a tercentennial look.
agglutinative features. Proc Mayo Clin 1935;10:609. Surgery 1968;64:685–700.
115. Ernstene AC, Gardner WJ: The effect of splanchnic nerve 131. Keynes G: Tercentenary of blood transfusion. Br Med J
resection and sympathetic ganglionectomy in a case of par- 1967;516:410–411.
oxysmal hemoglobinuria. J Clin Invest 1935;14:799. 132. Denis J: Phil Trans R Soc Lond [Biol] 1668;4:617.
116. Stats D, Wasserman LR: Cold hemagglutination: An interpre- 133. Walton MT: The first blood transfusion: French or English?
tive review. Medicine 1943;22:363. Med Hist 1974;18:360–364.
C H A P T E R 2

The Diagnosis of
Hemolytic Anemia

This chapter offers an approach However, before proceeding to a consideration of each


to the diagnosis of hemolytic of these three aspects of diagnosis, a review of some
anemia that enables the phy- definitions is appropriate.
sician to make an accurate
diagnosis without superfluous
steps. In addition to findings DEFINITIONS
elicited in the history and
physician examination, numer- Hemolysis. Hemolysis is defined as a reduction of
ous laboratory tests are avail- the RBC life span to less than the normal range of
able that are relevant to the 100 to 120 days.
diagnosis of hemolytic anemia. These tests include the Compensated Hemolytic Disease. A compensatory
examination of the blood and bone marrow, serum increase in erythropoiesis may be adequate to prevent
bilirubin, lactate dehydrogenase, serum haptoglobin, the development of anemia, thus resulting in compen-
methemalbumin, plasma hemoglobin, urinary hemo- sated hemolytic disease.2-4
globin and hemosiderin, direct and indirect antiglobu- Anemia with a Hemolytic Component. Studies of
lin (Coombs’) tests, cold agglutinin titer, determination RBC survival have shown that RBC life span is often
of the thermal amplitude and specificity of antibodies shortened in a number of chronic anemias. These
in the patient’s serum and eluate, Donath-Landsteiner include the anemias associated with disseminated
test, measurement of survival of radiolabeled red malignant disease, leukemias, lymphomas, renal
blood cells (RBCs), measurement of endogenous pro- insufficiency, liver disease, rheumatoid arthritis, and
duction of carbon monoxide, among others. The avail- the megaloblastic anemias. Thus, the mechanism of
ability of such a large array of laboratory tests makes the anemia in many patients includes a mild decrease
their optimal utilization a difficult task for the physi- in RBC survival, but is primarily due to inadequate
cian who encounters such a patient only occasionally.1 marrow production (relative marrow failure). It is cus-
To put some order into what is often a rather disor- tomary not to classify such patients as having
ganized approach, it is useful to think in terms of hemolytic anemia.
three separate steps: Hemolytic Anemia. The diagnosis of hemolytic
anemia is justified if a major mechanism in causation
1. Determining whether the patient’s anemia is due to of a patient’s anemia is a shortened RBC life span.
hemolysis Acquired Hemolytic Anemia. All hemolytic
2. Utilizing a limited number of clinical and labora- anemias except those caused by a hereditary defect
tory findings to develop a tentative diagnosis con- are referred to as acquired. Acquired hemolytic
cerning the cause of the hemolysis anemias do not necessarily have an immune patho-
3. Performing confirmatory tests to establish the genesis, and it is inappropriate to use this term as a
specific etiology synonym for autoimmune hemolytic anemia (AIHA).

33
34 Immune Hemolytic Anemias

RBC
Haptoglobin

Hb
Albumin

Hb

Hb

Methemalbumin Hb-HP
Complex

Hb Hemosiderin

RES

FIGURE 2-1. The destruction of red blood cells (RBCs) intravascularly results in the liberation of hemoglobin
(Hb) from the RBC. The Hb combines with haptoglobin (HP), and the HP-Hb complexes are rapidly
catabolized in the reticuloendothelial system (RES), resulting in low levels of serum Hp. Also, when Hb is
liberated into the plasma in large quantities, some of the heme combines with plasma albumin, resulting in
the formation of methemalbumin. When HP has been saturated and the level of plasma Hb exceeds the
renal threshold, Hb appears in the urine. Hemoglobinuria persisting for at least several days results in the
deposition in the renal tubules of hemosiderin derived from the breakdown of Hb. The hemosiderin may be
excreted in the urine, probably as a result of the desquamation of the renal tubular cells.

Hereditary Hemolytic Anemias. Hereditary hemo- such cases, hemoglobinemia and hemoglobinuria are
lytic anemias are caused by a hereditary abnormality, not found, but there is an increase in serum bilirubin
usually affecting the RBC intracellular contents or values and in bilirubin degradation products in the
membrane structure. urine and stool (Fig. 2-2).5
Congenital Hemolytic Anemias. Although the Distinguishing Extravascular and Intravascular
words congenital and hereditary are often used syn- Hemolysis. The distinction between extravascular and
onymously, congenital is defined as existing at birth, intravascular hemolysis is not precise. For example, a
regardless of causation. Thus, hemolytic disease of the low level of serum haptoglobin occurs as a result of
fetus and newborn is a congenital but not hereditary the rapid catabolism of complexes of serum haptoglo-
hemolytic anemia. bin with free hemoglobin in the plasma, the latter
Intravascular Hemolysis. In some diseases, such as characteristically resulting from intravascular hemol-
mechanical hemolytic anemia, paroxysmal nocturnal ysis (see Fig. 2-1). However, low levels of serum hap-
hemoglobinuria (PNH), and paroxysmal cold hemo- toglobin occur in essentially all hemolytic anemias.
globinuria (PCH), the destruction of RBCs appears to Similarly, serum bilirubin values may be elevated
take place primarily in the intravascular space with regardless of the mechanism of RBC destruction.8
release of free hemoglobin into the blood. The cardinal Indeed, in patients with brisk hemolysis, hemoglo-
features of intravascular hemolysis are hemoglobine- binemia (free hemoglobin in the plasma) and hemo-
mia and hemoglobinuria. Intravascular hemolysis of globinuria (hemoglobin in the urine) may occur in
at least several days’ duration results in hemosideri- hemolytic states in which the predominant mecha-
nuria. Even with minimal degrees of intravascular nism of hemolysis is extravascular, as in hemolysis
hemolysis, the serum haptoglobin level will be low or caused by Rh antibodies.
absent (Fig. 2-1).5-7 The major clinical value of the distinction of
Extravascular Hemolysis. In most hemolytic intravascular and extravascular hemolysis is that,
anemias, RBC destruction takes place predominantly when hemoglobinemia and hemoglobinuria are
in the cells of the reticuloendothelial system (RES). In present, certain causes of hemolytic anemia are much
The Diagnosis of Hemolytic Anemia 35

Hemoglobin
RETICULOENDOTHELIAL to
SYSTEM Bilirubin

FIGURE 2-2. The destruction of RBCs


extravascularly within the cells of the
reticuloendothelial system results in the
Bilirubin degradation of hemoglobin and the
VASCULAR Attached
COMPARTMENT production of bilirubin. The bilirubin passes
Albumin into the plasma, forms a loose complex with
albumin, and is taken up by the liver, where it
is conjugated with glucuronic acid.
Conjugated bilirubin gives a positive direct
van den Bergh test, whereas unconjugated
bilirubin gives a positive indirect reaction.
Conjugation of Only conjugated bilirubin is readily excreted
Bilirubin with into the urine. Conjugated bilirubin largely
Glucuronic Acid passes via the hepatic ducts to the intestine,
Liver where it is reduced to stercobilinogen and is
Urobilinogen excreted. Part of the stercobilinogen is
absorbed from the bowel and is reexcreted
by the liver (enterohepatic circulation). Some
absorbed stercobilinogen is excreted by the
kidneys as urobilinogen. In hemolytic anemia,
the increased catabolism of bilrubin
Enterohepatic characteristically results in increased
Bilirubin Circulation of concentrations of unconjugated bilirubin in
Glucuronide the plasma and increased concentrations of
Stercobilinogen
in Bile bilirubin degradation products in feces and
urine.

Stercobilinogen
in Intestines

more likely to be present (see later section). Also, these are: (1) a complete blood count (or serial blood counts)
findings indicate the likelihood that severe hemolysis with emphasis on the reticulocyte count and on red
is occurring, necessitating urgent diagnostic evalua- cell morphology in the peripheral blood film, (2) a
tion and initiation of appropriate therapy. serum bilirubin level, (3) a serum lactic dehydroge-
nase (LDH) level, and (4) serum haptoglobin value.
Two other clinical indicators of hemolysis that are
DETERMINATION OF THE HEMOLYTIC too often ignored are a significant drop in the hemo-
NATURE OF AN ANEMIA globin level without blood loss and the patient’s
transfusion requirement. These findings are particu-
A patient’s history and physical examination may larly important when the diagnostic implications of
suggest the possibility of a hemolytic anemia (symp- serum bilirubin, LDH, and haptoglobin values may be
toms of anemia, acholuric jaundice, splenomegaly), clouded because of alternative explanations for these
but usually the manifestations are nonspecific and the abnormalities, as may be true when immune hemoly-
initial work-up involves a laboratory evaluation of sis occurs in patients in the setting of bone marrow
anemia. Although the presence of hemolysis can be and solid organ transplantation, aggressive chemo-
determined by direct measurement of RBC life span, therapy, liver failure, and the like.
RBC survival studies are rarely necessary.9 Instead,
indirect indications of hemolysis are sought by tests The Blood Count
that yield evidence of increased hemoglobin break-
down and of bone marrow regeneration. These fea- Hemolytic anemia can usually be diagnosed or
tures are common to all hemolytic anemias, regardless excluded from consideration on the basis of routine
of their etiology. blood counts, and, indeed, this remains the single
In this era of extraordinarily sophisticated labora- most important means of doing so. A patient’s RBC
tory technology, it seems incongruous that a few volume can only be decreased rapidly by two mecha-
easily performed studies usually suffice to determine nisms: bleeding and hemolysis. (A cessation of RBC
whether a patient’s anemia is hemolytic in nature. The production in a patient with a normal RBC life span
most helpful of the readily available laboratory tests will only result in a decrease in RBC count of about 1%
36 Immune Hemolytic Anemias

per day, as the normal RBC life span is about 100 mean normal number of reticulocytes), whereas a 5%
days.) Hence, an acute drop in hemoglobin value reticulocyte count in a person with 1,000,000 RBCs/μL
without evidence of blood loss must alert the clinician indicates an absolute reticulocyte count of only
to a possible diagnosis of hemolysis. A common error 50,000/μL (not increased from the mean normal
is to attribute a marked drop in the level of hemoglo- number of reticulocytes). Thus, one must “correct” the
bin to bleeding when there is evidence for only reticulocyte count for the degree of anemia. (The term
minimal blood loss. For example, if the hemoglobin of “absolute reticulocyte count” has also been used for
a 70-kg man decreases rapidly from 12 to 9 g/dL, this the reticulocyte percentage corrected for the degree of
would require the loss of about 2.5 units of blood. A anemia, but here it is used to mean the total number
drop in hemoglobin of this magnitude cannot be of reticulocytes per microliter.)
explained on the basis of a weakly positive test for
gastrointestinal blood loss.
Also, when no significant blood loss is present, CORRECTED RETICULOCYTE COUNT
hemolysis is indicated by stable or falling hemoglobin The traditional means of correcting the reticulocyte
and hematocrit values in the face of increased RBC count for the degree of anemia has been to multiply
production (i.e., an increased reticulocyte count). the reticulocyte percentage by the ratio of the patient’s
hemoglobin, hematocrit, or RBC count to the mean
Reticulocytes* normal value of the same measurement.10 Convenient
numbers to use for the normal values are 15 g hemo-
RETICULOCYTE COUNT globin, 45% hematocrit, and 5,000,000 RBCs/μL. For
OR RETICULOCYTE PERCENTAGE example, the corrected reticulocyte count in a patient
An elevated reticulocyte count is a reflection of the with a reticulocyte percentage of 18% and a hema-
compensatory increase in erythropoiesis by the bone tocrit of 15 is 18% × 15/45, or 6%. The corrected retic-
marrow, and it is, therefore, an indirect indication of ulocyte count of 6% indicates that the number of
shortened RBC life span. Reticulocytes can be used as circulating reticulocytes is six times the normal
an indication of increased erythropoiesis and, with a number.
few simple calculations, the rate of increase of ery-
thropoiesis above normal can be estimated. In the RETICULOCYTE MATURATION TIME
steady state, the rate of increase in erythropoiesis can
serve as an estimate of the degree of shortening of the The correction of the reticulocyte count for anemia
RBC life span. Reticulocyte counts have usually been does not accurately reflect the extent of RBC produc-
reported as a percentage of circulating RBCs. In tion because of varying reticulocyte maturation times
normal persons, the reticulocyte count is approx- in anemic patients. In normal individuals, reticulo-
imately 0.5% to 1.5%. cytes found in the circulation are identifiable as retic-
ulocytes for about 1 day, whereas in patients with
anemia and increased erythropoietic stimulation by
ABSOLUTE RETICULOCYTE COUNT erythropoietin, the marrow maturation time is short-
One must think of reticulocytes in terms of the total or ened and the maturation time for reticulocytes circu-
“absolute” number of reticulocytes per microliter in lating in the peripheral blood becomes longer. The
order to avoid erroneous conclusions concerning the reticulocytes that are released early are called “shift”
extent of RBC production by the bone marrow. Using or “stress” reticulocytes and are recognizable on the
the value of 5,000,000/μL as a normal RBC count, a blood smear through a relative increase in size and
normal reticulocyte percentage of 0.5 to 1.5 represents basophilia.11 It is evident that if reticulocytes survive
an “absolute reticulocyte count” of 25,000 to as such in the peripheral blood for 3 days instead of
75,000 reticulocytes per microliter, or a mean value of the usual 1 day, the corrected reticulocyte percentage
50,000/μL. overestimates RBC production by a factor of 3.
A reticulocyte count of 5% might suggest that the Accordingly, to more accurately reflect the rate of ery-
number of reticulocytes is five times higher than thropoiesis relative to normal, a second correction
normal as the mean normal reticulocyte count is 1%, must be made on the basis of the circulating reticulo-
but determining the absolute reticulocyte count cyte maturation time.
makes it evident that this is not true if the patient is
anemic. For example, a 5% reticulocyte count in a CALCULATING CIRCULATING RETICULOCYTE
person who has 4,000,000 RBCs/μL indicates an MATURATION TIME
absolute reticulocyte count of 200,000/μL (4 times the
Estimates of the circulating reticulocyte maturation
time in patients with varying degrees of anemia have
*This section was written in collaboration with Elliot M. Landaw,
MD, PhD, Professor and Chairman, Department of Bio- been developed by Hillman12 and Hillman and
mathematics, University of California Los Angeles Medical Center, Finch.10,11,13 Hillman12 subjected two patients with
Los Angeles, California. hemochromatosis and six normal volunteers to pro-
The Diagnosis of Hemolytic Anemia 37

longed phlebotomy programs to lower hematocrit 14.0


values to levels of 27% to 32% or 25% to 30%. He made
measurements of plasma iron turnover to determine 13.0
erythroid production by the marrow and performed
12.0
twice daily reticulocyte counts. He also estimated RBC
production from the level of phlebotomy, because, if 11.0
the hematocrit is kept at a constant level, red cell pro-
duction must equal the amount of packed red cells 10.0

Reticulocyte Count (%)


removed by phlebotomy plus the amount of red cells
dying each day. 9.0
A progressive shortening of the iron transit time
8.0
occurred as the severity of the anemia increased. By
the time the hematocrit was reduced to 25% to 30%, 7.0
the marrow iron transit time had shortened to
1.5 days, as compared to a nomal time of 3.5 days. 6.0
This was associated with the appearance in circulation
of large, polychromatic reticulocytes containing 5.0
greater than normal amounts of reticulum. These
4.0
findings implied a premature delivery into circulation
of marrow reticulocytes, which require a longer than 3.0
normal period of maturation in circulation to lose
their reticulum (the “circulating reticulocyte matura- 2.0
tion time”). Indeed, estimates of marrow RBC produc-
tion from absolute reticulocyte counts exceeded other 1.0
indices of RBC production by a factor of 1.5 to 3.0 as
production increased from normal to two to five times 1.0 2.0 3.0 4.0 5.0 6.0 7.0 8.0
normal12 (Fig. 2-3). Marrow Production (⫻ normal)
Circulating reticulocyte maturation times were cal- FIGURE 2-3. A comparison of the absolute reticulocyte counts and
culated from the absolute reticulocyte counts and plasma iron turnover (closed circles) or phlebotomy production indices
plasma iron turnover production indices at varying (open circles) demonstrated a poor correlation. The absolute
levels of marrow production and hematocrit depres- reticulocyte count exceeded marrow production by one and one-half to
three times normal over a wide range of production levels. (From
sion. As shown in Figure 2-4, when these circulating Hillman RS: Characteristics of marrow production and reticulocyte
reticulocyte maturation times were compared to the maturation in normal man in response to anemia. J Clin Invest
hematocrit level, they progressively lengthened with 1969;48:443–453.)
increasing anemia. From a normal value of 0.8 to
1.2 days, the circulating reticulocyte maturation time CALCULATING THE RETICULOCYTE
lengthened to 1.5 to 2 days at hematocrit levels of 32% PRODUCTION INDEX
to 37% and to 1.7 to 3 days at hematocrit levels of 25%
to 30%. Hillman concluded that only by correcting the Using the second correction of the reticulocyte count,
absolute reticulocyte count for the appropriate prolon- which takes into account the circulating reticulocyte
gation of circulating reticulocyte maturation time can a maturation time, one can now calculate the “reticulo-
reasonable index of RBC production be obtained.12 cyte production index,” which estimates the rate of
Further, it is possible to estimate a maturation time for erythropoiesis relative to normal. For example, con-
a specific level of hematocrit depression in those states sider a patient whose reticulocyte percentage is 15%
in which erythropoietin stimulation appears to be and whose hematocrit level is 25%. The corrected
appropriate for the severity of anemia. Thus, a correc- reticulocyte count is 15% × 25/45, or 8%. The circulat-
tion factor can be derived according to the hematocrit ing reticulocyte maturation time correction factor
level, and this factor is then used to carry out a second when the hematocrit is 25% is 2, and the reticulocyte
correction of the reticulocyte count. production index is 8/2, or 4. Therefore, in this patient
Based on the data of Hillman12 and Hillman and with a reticulocyte count of 15%, erythropoiesis is
Finch,11,13 it is appropriate to apply a second correc- estimated to be four times the normal rate.
tion factor to the corrected reticulocyte count to One may also derive the reticulocyte production
account for varying circulating (blood) reticulocyte index by dividing the absolute reticulocyte count by
maturation times. One does this by dividing the cor- the circulating reticulocyte maturation time factor and
rected reticulocyte count by the following circulating comparing this value to the mean normal absolute
reticulocyte maturation time correction factors: hema- reticulocyte count of 50,000/μL. Thus, if the absolute
tocrit of 40 to 45, correction factor 1.0; hematocrit of reticulocyte count is 400,000 in a patient with a hema-
35 to 40, correction factor 1.5; hematocrit of 25 to 35, tocrit value of 26 (circulating reticulocyte maturation
correction factor 2.0; hematocrit of less than 15 to 25, correction factor = 2.0), the corrected absolute reticu-
correction factor 2.5 (Fig. 2-5). locyte count is 200,000. This is to be compared with
38 Immune Hemolytic Anemias

3.0 tion and destruction are equal. A rough estimate of the


mean RBC life span may be made simply from the
reticulocyte production index. If this value is, for
example, five times normal, the mean RBC life span is
one fifth of normal, or 20 days, if one uses 100 days as
Reticulocyte Maturation Time (days)

2.5
the normal mean RBC life span. The mean RBC life
span can be calculated from the reticulocyte produc-
tion index by multiplying the reciprocal of the pro-
duction index by the mean normal life span. In this
2.0 example,

RBC survival = 1 × 100 = 20 days


5

1.5
Chen and Wang14 used the data of Hillman12 to
develop a slightly more detailed means of calculating
the circulating reticulocyte maturation time [matura-
tion time in days = 3.25 – (0.05 × hematocrit)]. They
1.0 compared published values for mean erythrocyte life
span, as measured by DF32P (diisopropylfluorophos-
phate), in 43 patients with chronic hemolytic anemia
due to sickle cell disease15,16 with those calculated
45 40 35 30 25 from the reticulocyte count and hematocrit levels. The
Hematocrit (%) calculated hemolytic rate correlated significantly with
FIGURE 2-4. Progressive prolongation of the circulation reticulocyte the measured rate (p < 0.001). Using a small correction
maturation time occurred with depression of the hematocrit. If we factor to correct for the enrichment in the circulation
assume that the discrepancy between the absolute reticulocyte count of RBCs with lower hemolytic rates, the calculated
and plasma iron turnover is primarily related to a premature delivery
of marrow reticulocytes, which then require a longer time to lose their and measured hemolytic rates were almost identical.
reticulum, the reticulocyte maturation time was calculated as: These results validated the studies of Hillman12
regarding methods for estimating circulating reticulo-
absolute reticulocyte count cyte maturation times.
Reticulocyte maturation time (days) =
production index Chen and Wang14 have concluded that a simple
Closed circles, calculated from the plasma iron turnover; open circles,
mathematical model using hematocrit levels and
calculated from phlebotomy production measurements. (From Hillman reticulocyte counts provides reasonable estimates of
RS: Characteristics of marrow production and reticulocyte maturation the mean steady-state hemolytic rate in patients with
in normal man in response to anemia. J Clin Invest 1969;48:443–453.) chronic hemolysis. Their formula for determining the
hemolytic rate (the percentage of RBC turnover per
day), k, is

k = –ln (1 – r)/λ × 100%


the mean normal absolute reticulocyte count of
50,000/μL. The reticulocyte production index is, In this formula, r = the reticulocyte fraction, and
therefore, 200,000/50,000 or 4. λ = the reticulocyte maturation time in days, which is
The use of the circulating reticulocyte maturation calculated as described in the equation [maturation
time correction factor is only appropriate when there time in days = 3.25 – (0.05 × hematocrit)]. The mean
are readily apparent polychromatophilic macrocytes RBC production index is 1/k. For example:
on the peripheral blood smear, indicating the presence For a patient with a hematocrit of 25% and a reticulocyte
of shift cells. Such calculations do not apply if ery- count of 20%:
thropoiesis is suppressed, as by infection, in which
case reticulocyte maturation time is variable.
Reticulocyte maturation time
= 3.25 – (0.05 × 25) = 3.25 – 1.25 = 2.0 days
RBC LIFE SPAN
Calculations of the reticulocyte production index can
–ln(1 – 0.20)
be used to provide a reasonable estimate of the ery- Hemolytic rate = k = × 100% = 11.2%/day
throcyte life span in patients with hemolytic anemia 2
who are in steady state, as evidenced by a stable
hematocrit level and reticulocyte count. This is true Production index = 11.2 × 25 = 6.2
because, in the steady state, the rate of RBC produc- 45
The Diagnosis of Hemolytic Anemia 39

Marrow
FIGURE 2-5. Correlation of the hematocrit with the
Normoblasts and Blood
marrow and blood reticulocyte maturation times.
Reticulocytes Reticulocytes Ordinarily, erythropoietin increases in proportion to the
Hematocrit Maturation Time (Days) Maturation Time (Days) degree of anemia present. With increasing erythro-
poietin stimulation, the maturation time of the erythroid
marrow normoblasts and marrow reticulocytes
45 3.5 1.0 progressively shortens from a normal of 3.5 days to as
little as 1.5 to 1.0 day. Much of this shortening is
secondary to a shift of marrow reticulocytes into the
circulation. This results in a prolongation of the
35 3.0 1.5 maturation time of circulating blood reticulocytes from
a normal of 1 day to as much as 2.5 to 3.0 days with
severe anemia. This needs to be taken into account
when calculating the reticlocyte production index. The
25 2.5 2.0 maturation time values shown for the blood
reticulocytes can be used as a correction factor in this
calculation (see text). (From Hillman RS, Finch CA: The
detection of anemia. In Red Cell Manual, Philadelphia:
15 1.5 2.5 F. A. Davis, 1969:39–65.

1 is due to the application of individual criteria in retic-


RBC survival =
production index ulocyte identification. Although careful counting of an
adequate sample19 by individual technologists and
× mean normal life span in days the use of an ocular insert20 can result in clinically
useful information, development of automated reticu-
= 1 = (0.16) (100) = 16 days locyte analysis has resulted in increased accuracy.
6.2 Automated reticulocyte analysis has its basis in the
technology of flow cytometry.21 A number of instru-
ments have been developed by various manufactur-
ESTIMATING RBC LIFE SPAN ers, and they all provide improved precision over
USING NOMOGRAMS manual microscopic techniques.
Using the hematocrit and the reticulocyte percentage,
one can determine the hemolytic rate constant from RETICULOCYTE MATURATION INDEX
the nomogram in Figure 2-6. Using the hematocrit and
the absolute reticulocyte count, one may determine Some investigators have proposed the use of a “retic-
the production index from the nomogram in ulocyte maturity index” (RMI) to indicate the presence
Figure 2-7. From these values, RBC survival can then of “shift” reticulocytes. The RMI can be calculated
be calculated as indicated earlier. either by mean fluorescence intensity measurements
Another evaluation of the validity of using reticu- of the reticulocyte population or by deriving a frac-
locyte counts to quantify RBC production was tional index based on the relative proportion of the
reported by Rhyner and Ganzoni,17 who determined highly fluorescent reticulocyte fraction of the total
effective erythroid marrow activity by iron kinetic population.22 There is an inverse relationship between
studies and compared these results to values the hemoglobin or hematocrit and the RMI, as would
obtained using reticulocyte counts. They confirmed be expected from the data of Hillman and Finch.11
that reticulocyte counts must be corrected for Unfortunately, no studies have been done comparing
anemia and circulating reticulocyte maturation red cell production or life span as determined using
times as just described. Using these corrections, they radiolabeled RBC survival determinations or iron
found excellent agreement between the two kinetic studies with calculations based on the RMI.
methods for assessing effective erythropoiesis in Further, there has been no standardization of the RMI
both normal individuals and in the 11 patients they between laboratories, and the evaluations of the clini-
studied who had acquired hemolytic anemias cal significance of the RMI in anemic patients has
(Fig. 2-8; Table 2-1). included data on very few patients with hemolytic
anemia.23,24
ACCURACY OF RETICULOCYTE COUNTING
DIRECT DETERMINATION OF THE ABSOLUTE
An obstacle to the determination of the reticulocyte RETICULOCYTE COUNT
production index and mean erythrocyte life span from
reticulocyte counts and the hematocrit is the lack Flow cytometers directly determine the absolute retic-
of precision of the manual reticulocyte count.18 ulocyte count, thus making unnecessary the calcula-
Technologist-to-technologist variation is the major tion of this value from the RBC and reticulocyte
source of inaccuracy at all reticulocyte levels, and this percentage. In some of the methods described for flow
40 Immune Hemolytic Anemias

45
Hemolytic
Rate Constant
(% per day)
40

20

35
15

12

Hematocrit (%)
30
10
FIGURE 2-6. Relationship between hematocrit and 8
reticulocyte percentages at various hemolytic rate constants. 7
25
6
5
4
20
3
2
1
15

10
0 5 10 15 20 25 30
Reticulocyte Percentage

45
Production Index
10 (normal = 1)
40
8
7
35
6

5
Hematocrit (%)

30
4
FIGURE 2-7. Relationship between hematocrit and absolute
reticulocyte count at various red blood cell production indices.
25
3
2.5
20
2
1.5
1
0.5
15

10
0 1 2 3 4 5 6 7 8 9
Absolute Reticulocyte Count (×100,000/ μ L)

cytometric analysis of reticulocytes, the analysis is than is true for the more reproducible automated
adjusted to exclude counting of the “stress” or “shift” reticulocyte counts.
reticulocytes. Again, there are no data validating this
adjustment by comparison with iron kinetic data or INTERPRETATION OF AN ELEVATED
RBC survival studies, as has been done with manually RETICULOCYTE COUNT
determined reticulocyte counts. Ironically, then, there
is more information relating manually determined A simple but surprisingly accurate “rule of thumb” is
reticulocyte counts to RBC production and survival that one should suspect hemolytic anemia whenever
The Diagnosis of Hemolytic Anemia 41

FIGURE 2-8. Comparison between effective RBC production


Production Index, R

4 as evaluated by ferrokinetic measurements (EIT) and


reticulocyte counts (R). The EIT found in patients was divided
by 0.49 to obtain the production index EIT, where 0.49
corresponds to mg iron utilized for RBC production per day
3 in normal individuals. Reticulocyte counts were corrected for
hematocrit and for marrow transit time. (From Rhyner K,
Ganzoni A: Erythrokinetics: Evaluation of red cell production
by ferrokinetics and reticulocyte counts. Eur J Clin Invest
2 1972;2:96–101.)
r = 0.93
r = 0.93x – 0.07

0 1 2 3 4 5 6 7
Production Index, EIT

TABLE 2-1. COMPARISON OF CALCULATED AND MEASURED ERYTHROCYTE PRODUCTION INDICES

Age Hematocrit Reticulocyte Marrow Transit Production Production


Subject (yr) Sex Diagnosis (%) Count (%) Time (days) Index (Retic)* Index (EIT)†

1 45 M Normal 50.0 0.9 3.5 1.00 1.08


2 69 M Normal 40.0 1.4 3.5 1.12 1.30
3 39 M Normal 40.0 0.9 3.5 0.76 0.79
4 39 M Normal 45.0 0.8 3.5 0.80 0.85
5 34 M Normal 50.0 1.3 3.5 1.78 1.21
6 22 M Normal 42.0 1.0 3.4 0.90 0.76
Mean 45.0 1.0 3.5 1.06 1.00
SD 4.6 0.2 0.04 0.38 0.23

24 55 M Hemolytic anemia 31.0 7.8 1.8 2.76 2.65


25 60 F Hemolytic anemia 30.0 4.7 2.7 2.32 1.94
26 70 F Hemolytic anemia 36.0 9.6 2.0 4.38 3.47
27 64 F Hemolytic anemia 33.0 5.7 2.9 3.47 3.81
28 31 M Hemolytic anemia 38.0 3.9 3.3 3.11 3.38
29 75 F Hemolytic anemia 16.5 29.0 2.1 6.36 6.57
30 60 F Hemolytic anemia 18.0 32.0 1.4 5.10 5.22
31 25 F Hemolytic anemia 20.0 8.3 2.3 2.42 2.57
32 38 M Hemolytic anemia 35.0 3.9 2.6 2.25 2.45
33 53 M Hemolytic anemia 33.5 3.7 3.0 2.36 2.04
34 63 F Hemolytic anemia 28.0 5.8 2.2 2.26 2.06

EIT, erythrocyte iron turnover.


* Production index calculated using corrected reticulocyte counts.
† Production index measured by erythrocyte iron turnover.
Data from Rhyner K, Ganzoni A: Erythrokinetics: Evaluation of red cell production by ferrokinetics and reticulocyte counts. Eur J Clin Invest 1972;2:96–101.

the uncorrected reticulocyte count is greater than 5%. An elevated reticulocyte production index is a more
The probability of hemolytic anemia rapidly increases informative indication of the degree of increased ery-
with increasing degrees of reticulocytosis and, if the thropoiesis. Within a few days of the development of
uncorrected reticulocyte count is greater than 10%, the moderate anemia, RBC production in the marrow
diagnosis is very likely. may increase to a level three to five times normal.
42 Immune Hemolytic Anemias

Under conditions of prolonged stimulation, patients Bilirubin


with hemolytic anemia may have rates of 6 to 10 times
normal.11 For patients in the steady state, a reasonable Bilirubin is formed mainly in the RES system by enzy-
estimate of RBC survival can be made from the reticu- matic degradation of hemoglobin from senescent
locyte count and hematocrit level, as described earlier. RBCs.25 About 15%, the so-called early labeled pigment
Although an elevated reticulocyte count accurately or shunt bilirubin, is produced in the liver from
indicates the presence or absence of hemolytic anemia nonhemoglobin heme, such as the cytochromes, or in
in a surprisingly high percentage of patients, excep- the bone marrow from RBC precursors, as in
tions do, of course, occur. A reticulocytosis can occur intramedullary hemolysis or ineffective erythropoiesis.
for other reasons, such as blood loss or recent treat- Unconjugated bilirubin in the serum is bound to
ment of megaloblastic anemia. However, blood loss albumin and is transported to the liver, where it
causes a problem in differential diagnosis rather is taken up by hepatic acceptor protein. The hepatic
infrequently because sustained bleeding that is of microsomal enzyme, glucuronyl transferase, trans-
sufficient volume and duration to result in a reticulo- forms unconjugated bilirubin to water-soluble conju-
cyte count high enough to cause a strong suspicion of gated bilirubin, primarily bilirubin diglucuronide,
hemolysis can result only from clinically evident which is then excreted in the bile.10 In general, the con-
blood loss. Similarly, confusing treated megaloblastic jugated bilirubin is measured by the direct reacting
anemia with hemolytic anemia is an infrequent clini- fraction and the unconjugated bilirubin by the indirect
cal problem. In contrast to megaloblastic anemia, reacting fraction. Hyperbilirubinemias of unconjugated
treatment of iron deficiency anemia generally pro- bilirubin are referred to as acholuric jaundice because
duces only a modest reticulocyte response.8 unconjugated bilirubin cannot be excreted in the urine.
In cases in which there is doubt, repeat blood counts In conjugated hyperbilirubinemias a small fraction of
will be of value as a persistently elevated absolute serum conjugated bilirubin is excreted in the urine.26
reticulocyte count and reticulocyte production index Hyperbilirubinemia is usual in hemolytic anemia,
without an increase in hemoglobin and in the absence but it is not a constant finding, so its absence does not
of blood loss is diagnostic of hemolysis. exclude the diagnosis. The indirect reacting fraction is
the predominant fraction elevated in the presence of
hemolysis. The direct reacting fraction is characteristi-
RETICULOCYTOPENIA IN PATIENTS cally elevated in conditions in which true plasma
WITH HEMOLYTIC ANEMIA conjugated hyperbilirubinemia results from a reflux of
conjugated bilirubin from the liver or biliary tract
Reticulocytopenia in the presence of hemolytic to the blood. Conjugated hyperbilirubinemia indicates
anemia presents a difficult problem. It is discussed in either a physiologic or mechanical obstruction to the
more depth in Chapter 3. flow of bile, which may be located at any point from
If the hemolysis is of abrupt onset, at least several within the hepatocyte itself to the duodenum.
days must elapse before the development of a reticulo- Hepatocellular injury of any type substantially reduces
cytosis. Patients with hemolytic anemia that is less the capacity of the hepatocyte to transport bilirubin
acute in onset may also have reticulocytopenia because into bile. Hence, hemolysis ocurring in the setting of
of bone marrow suppression for various reasons, liver disease frequently results in a combination of con-
including autoantibody reactivity against erythroid jugated as well as unconjugated hyperbilirubinemia.27
precursors. Although the initial reticulocyte count may The plasma concentration of indirect reacting
misleadingly suggest that hemolytic anemia is not (unconjugated) bilirubin is 0.2 to 0.9 mg/dL for 95% of
present, the diagnosis of hemolysis can nevertheless be a normal population, and 99% of such a population
made simply on the basis of the blood count if serial will have a value less than 1.0 mg/dL.27 The upper
determinations are made over a period of several days limit of normal for direct reacting (conjugated) biliru-
or more. This is true because the combination of bin in the presence of a normal total plasma bilirubin
hemolysis and reticulocytopenia results in a rapidly concentration (<1.2 mg/dL) is 0.2 mg/dL.
falling hemoglobin and hematocrit level, which can Plasma bilirubin turnover studies have indicated
result only from hemolysis, provided significant blood that, in patients with chronic hemolysis who are in the
loss is excluded. steady state, unconjugated bilirubin concentrations in
excess of 4 mg/dL do not result from hemolysis per
RBC Morphology se, but imply a concomitant reduction in hepatic func-
tion. This calculation is based on an assumed
The RBC morphology in the peripheral blood film not maximum increase in RBC production by the bone
only frequently substantiates the impression of hemo- marrow to a rate of eight times the normal rate. This
lysis, but often suggests a specific diagnosis or a observation does not apply to acute hemolytic
limited number of diagnostic possibilities. Morpholo- episodes, which are non–steady-state situations, in
gic findings are discussed in more detail later (see which case the rate of RBC destruction and bilirubin
page 53). production may be greater.
The Diagnosis of Hemolytic Anemia 43

Many patients with low-grade hemolytic anemia BILIRUBIN VALUES IN PATIENTS


will be found to have unconjugated bilirubin con- WITH HEMOLYTIC ANEMIAS
centrations of less than 1 mg/dL, which are con-
ventionally interpreted as normal. However, Berlin Tisdale and coworkers35 performed 77 bilirubin deter-
and Berk27 point out that unconjugated bilirubin minations in 46 patients with hemolytic anemia and
levels must be corrected for anemia in a manner found that the direct-reacting fraction constituted less
analogous to correcting reticulocyte percentages for than 15% of the total in most patients, especially if the
anemia. They suggest correcting the upper limit of total bilirubin was 4.0 mg/dL or greater. The
normal for the serum unconjugated bilirubin as direct-reacting fraction exceeded 1.2 mg/dL in only
follows: five instances unless there was definite evidence of
concomitant hepatic disease.
Pirofsky36 reported that an elevation in serum
Upper limit of normal = 1.0 mg/dL
bilirubin value was one of the most consistent abnor-
patient’s hematocrit
× malities noted in patients with AIHA. Total serum
45 bilirubin was determined in 120 patients. An elevated
value was found in 66 cases, or 55% of the tested
patients. In 57 patients, the indirect bilirubin was
CLINICAL INTERPRETATION OF UNCONJUGATED increased above normal. In 20 patients, the increased
HYPERBILIRUBINEMIA bilirubin value was exclusively of an indirect variety.
Allgood and Chaplin37 found that the indirect-
Chronic unconjugated hyperbilirubinemia can be reacting bilirubin value was less than 1.0 mg/dL in
attributable to increased plasma bilirubin turnover, to only 13% of 47 patients with AIHA. It varied from less
reduced hepatic bilirubin clearance, or to some than 0.8 to 8.6 mg/dL, and in 9 of the 47 patients, it
combination of the two. Hemolysis is the only source was greater than 4 mg/dL. The direct-reacting frac-
of increased bilirubin turnover in humans. Reduced tion was significantly elevated in only five patients,
hepatic bilirubin clearance associated with structural and the highest value was 3.6 mg/dL. In two of these
liver disease is almost inevitably accompanied by patients, there was definite evidence of underlying
other biochemical evidence of hepatic dysfunction, liver disease; in the other three, it was suspected.
including elevated levels of direct bilirubin. An excep- Although indirect-reacting hyperbilirubinemia is
tion to this generalization is Gilbert’s syndrome,27-29 characteristic of hemolytic anemia, some authors have
which is an inherited form of mild unconjugated reported that an increase in conjugated (direct-
hyperbilirubinemia characterized by decreased biliru- reacting) bilirubin does occur in a minority of patients,
bin UDP-glucuronyl transferase activity (UGTA1).30 even in the absence of liver dysfunction.26,35,36,38 Even
Homozygosity for the variant promoter for the less well recognized is the fact that bilirubinuria is
UGTA1 gene, which is associated with Gilbert’s syn- seen occasionally in patients with uncomplicated
drome, is frequently encountered in European and hemolytic jaundice.35,38
African populations with a prevalence of 11% and
19%, respectively.31
The greatest importance of the serum bilirubin
OTHER ASPECTS OF BILIRUBIN METABOLISM
level in regard to the diagnosis of hemolytic anemia
lies in the fact that, if a patient has a significantly ele- Within the gastrointestinal tract, bilirubin is further
vated serum bilirubin level (characteristically, but degraded by bacterial action to a series of urobilino-
not invariably, between 1 and 4 mg/dL), which is gens and a number of other products. Although the
almost exclusively of the unconjugated or indirect conversion of heme to bilirubin and carbon monoxide
reacting type, the presence of hemolysis (or Gilbert’s is quantitative, the further degradation of bilirubin to
syndrome) is almost assured. In other disorders urobilinogens is not. Hence, measurements of fecal
with unconjugated hyperbilirubinemia, such as tha- urobilinogen excretion may appreciably underesti-
lassemia,32 some cases of sideroblastic anemia,33 mate total heme degradation.27 Although raised
dyserythropoietic jaundice,34 megaloblastic anemia, values are considered to be valid indications of hemo-
and ineffective erythropoiesis,27 the hyperbilirubine- lysis, the tests are difficult to perform accurately and
mia is due, at least in part, to hemolysis, including are usually not necessary.
destruction of erythroid precursors in the bone
marrow.
Changes in plasma unconjugated bilirubin concen-
tration over time can be an extremely useful indicator
Serum Haptoglobin
of altered physiology. A decline in the plasma uncon- Haptoglobin is a dimeric glycoprotein comprising
jugated bilirubin concentration provides early evi- two α chains and two β chains that bind to hemo-
dence of a decrease in the rate of hemolysis in a globin α dimers. The normal range of serum hapto-
patient with hemolytic anemia.27 globin is about 50 to 150 mg/dL. When RBCs are
44 Immune Hemolytic Anemias

destroyed in the vascular compartment, the hemo- hypersplenism had normal values of serum haptoglo-
globin escaping into the plasma is bound to hapto- bin, and these were considered false-negative results.)
globin. The haptoglobin-hemoglobin complex is Shinton and coworkers42 found the range of serum
cleared from the plasma with a half-life (T1⁄2) of 10 to haptoglobin in 110 normal subjects to be 33 to
30 minutes.39 In contrast, free haptoglobin has a 213 mg/dL; 80% of patients with hemolytic disease or
half-life of 5 days, so that when large amounts of megaloblastic anemia had subnormal levels.
the rapidly turned over haptoglobin-hemoglobin Subnormal levels were also found in some patients
complex are formed, the haptoglobin content of the with hemorrhage into the tissues and occasionally in
plasma is depleted. Haptoglobin is diminished not association with other diseases. They concluded that,
only in the plasma of patients undergoing frank when taken in conjunction with other clinical and lab-
intravascular hemolysis, but also in the plasma of oratory features, serum haptoglobin measurements
patients who have accelerated red cell destruction that can be of diagnostic value.
occurs primarily within macrophages. Presumably, The serum haptoglobin value after infusion of
there is either enough intravascular hemolysis in such hemoglobin returns to about half the initial value
hemolytic disorders to lower the plasma haptoglobin within 36 hours. Subsequent increments return the
level or enough leakage from the phagocytic cells into concentration nearly to preinfusion levels after 6 to
the plasma to bind to haptoglobin. Thus the measure- 10 days. This rapid recovery of circulating haptoglo-
ment of haptoglobin in plasma or serum has some bin is of clinical importance, as within 2 days of a tran-
usefulness in diagnosing the presence of hemolysis.39 sient hemolytic episode, plasma haptoglobin levels
may no longer be sufficiently low to be indicative of
accelerated red cell destruction.43
SENSITIVITY IN DEMONSTRATION OF HEMOLYSIS
When hemoglobin destruction exceeds two or three PATIENTS WITH PROSTHETIC HEART VALVES
times the normal rate, the serum haptoglobin level
will usually be low,7 and even less hemolysis is neces- Cullhed44 measured serum haptoglobin in 26
sary to result in low serum haptoglobin levels if the patients with nonoperated aortic valve disease, in
hemolysis is primarily intravascular. Indeed, a very 15 patients with a Starr-Edwards aortic prosthesis, in
small degree of intravascular hemolysis, insufficient 10 patients with a similar mitral prosthesis, and in 1
to increase the daily turnover of hemoglobin appre- patient with a double aortic and mitral prosthesis. In
ciably above normal, can deplete the serum com- most operated cases, the haptoglobin level was deter-
pletely of haptoglobin.7 Thus, the level of serum mined 1 year after the operation. Normal values were
haptoglobin can be considered a reasonably sensitive found in all but two of the nonoperated aortic cases,
indicator of hemolysis, although it must be kept in whereas a low value was the rule in cases with aortic
mind that even greater rates of hemoglobin break- or mitral ball-valve prostheses. The hemoglobin and
down may be necessary to depress the level in situa- hematocrit values were normal in all operated
tions in which there is apparently an increased rate of patients, and a modest reticulocytosis (2%–4%) was
synthesis, such as in acute or chronic inflammatory present in only four of the patients with aortic pros-
disease, neoplasia (including lymphomas), or steroid theses. Slightly increased serum bilirubin concentra-
administration. Also, in severe hepatocellular disease, tions (1.4 and 1.6 mg/dL, respectively) were found in
the serum haptoglobin level may be low, probably as two aortic cases. The findings of these authors are con-
a result of decreased production. Congenital hapto- sistent with the data of Brus and Lewis7 indicating
globin deficiency also occurs, but reports of this that small amounts of primarily intravascular hemol-
disorder are rare.40 ysis can deplete the serum of haptoglobin.

CLINICAL EXPERIENCES HAPTOGLOBIN LEVELS FOLLOWING


BLOOD TRANSFUSION
Marchand and coworkers41 performed haptoglobin
assays on 100 patients with a variety of hematologic Two groups of investigators studied the effects of blood
and nonhematologic conditions to determine the use- transfusion on serum haptoglobin levels. Langley and
fulness of serum haptoglobin in the diagnosis of coworkers45 studied changes in serum haptoglobin
hemolysis. They found that values of less than 25 values in 21 patients following 29 transfusions of RBC
mg/dL, as an indication of hemolysis, provided sensi- stored for an average of 7.1 days. They found that trans-
tivity and specificity of 83% and 96%, respectively, fusion produced little effect on the serum haptoglobin.
and that the predictive value for hemolytic disease In only three instances was a decrease of more than
was 87% (Table 2-2; Fig. 2-9). However, their study 25 mg/dL observed. Fink and coworkers46 measured
population included only 10 cases of AIHA, 6 cases of serum haptoglobin following transfusion of 356 units of
mechanical hemolysis, 6 cases of megaloblastic RBCs in 120 patients, and also measured serum hapto-
anemia, and 2 cases of hypersplenism. (Two patients globin in 65 patients who were diagnosed as having had
with megaloblastic anemia and the two patients with a febrile and/or allergic nonhemolytic transfusion reac-
The Diagnosis of Hemolytic Anemia 45

TABLE 2-2. SCREENING FOR HEMOLYSIS WITH A HAPTOGLOBIN CUTOFF OF 25 MG/DL*

No. of Patients with No. of Patients with


Haptoglobin ≤25 mg/dL Haptoglobin >25 mg/dL Total

Hemolysis 20 4 24
No hemolysis 3 73 76
Total 23 77 100

* Sensitivity, 83%; specificity, 96%; efficiency, 93%; predictive value of positive result, 87%; and predictive value of negative result,
95%.
From Marchand A, Galen RS, Van Lente F: The predictive value of serum haptoglobin in hemolytic disease. JAMA 1980;243:1909–1911.

tion. In the entire series of 185 patients, there were only Serum Lactic Dehydrogenase
four cases in which the pretransfusion level was within
normal limits and the post-transfusion level was less Serum lactic dehydrogenase (LDH) has also been
than 30 mg/dL. In all four cases, clinical information utilized in the diagnosis of hemolytic anemia because
adequately explained the drop in serum haptoglobin, RBCs have a high content of the enzyme. Stein47
that is, absorption of hemoglobin into the plasma from studied LDH activity in patients with hemolytic
a large hematoma. Because low levels of serum hapto- anemia, including patients with primarily intravascu-
globin have been documented following hemolytic lar hemolysis (e.g., those with prosthetic heart valves
transfusion reactions,45,46 serum haptoglobin measure- PNH) or primarily extravascular hemolysis (e.g.,
ment may be considered a diagnostic aid when evaluat- those with hereditary spherocytosis) (Table 2-3). The
ing a patient for a possible hemolytic transfusion degree of elevation in hereditary spherocytosis was
reaction. modest even with severe hemolysis, although
intravascular hemolysis always produced a marked
elevation.
Myhre and coworkers48 performed RBC survival
+ 450 studies with 51Cr-labeled RBCs in patients with
various LDH levels several months after insertion of
ball-valve aortic and/or mitral prostheses. They
400 found a close correlation between LDH and RBC sur-
vival, suggesting that LDH level is a reliable parame-
ter of the degree of intravascular hemolysis (Fig. 2-10).
350 They also used published data from a series of 50
patients with ball-valve prostheses49 and found that
48 of the 50 observations fell within the 95%
300
confidence interval of their regression line (Fig. 2-11).
Haptoglobin, mg/dL

From these data, the authors suggested that it is pos-


250 sible to approximate the erythrocyte destruction rate
from LDH levels for patients with intravascular
hemolysis, as indicated in Table 2-4. Similar correla-
200 tions have not been made in hemolytic disorders with
primarily extravascular hemolysis.
An analysis of the sensitivity, specificity, predictive
150 value, and efficiency of LDH and haptoglobin alone or
in combination in the diagnosis of hemolysis41 indi-
cates that haptoglobin and LDH each had a sensitivity
100 of 83%. Requiring either test to be positive yielded a
sensitivity of 92%, but for LDH alone, the specificity
was only 61% and the predictive value only 40%.
50 Because LDH is an inexpensive and commonly per-
25 formed test and has a high degree of sensitivity, it is a
good screening test for the evaluation of hemolysis,
Hemolytic Nonhemolytic provided other common causes of an increase are
excluded. Isoenzyme fractionation of the elevated
FIGURE 2-9. Serum haptoglobin levels in hemolytic and nonhemolytic
disorders. (From Marchand A, Galen RS, Van Lente F: The predictive
LDH in hemolysis may demonstrate increased LD1
value of serum haptoglobin in hemolytic disease. JAMA levels that are out of proportion to the LD2 fraction.50
1980;243:1909–1911.) Serial determinations of LDH can be performed to
46 Immune Hemolytic Anemias

TABLE 2-3. SERUM LACTIC DEHYDROGENASE IN HEMOLYTIC ANEMIA

Reticulocyte
PCV Count Serum LDH
Patient Diagnosis (%) (%) (n = 250–800 U/mL)

1 Three artificial heart valves 27 7.4 5,040


2 Two artificial heart valves 27 10.4 4,200
3 One artificial heart valve 25 10.0 8,250
4 Paroxysmal nocturnal 24 9.0 9,720
hemoglobinuria
5 Paroxysmal nocturnal 12 22.4 22,800
hemoglobinuria
6 Paroxysmal nocturnal 27 8.2 8,520
hemoglobinuria
7 Acid ingestion 40 2.1 4,960
8 Chemical abortion 19 3.7 4,400
9 Burns 31 3.0 9,390
10 Vasculitis 27 7.4 7,020
11 Hereditary spherocytosis 28 6.8 700
12 Hereditary spherocytosis 29 10.0 450
13 Hereditary spherocytosis 21 28.4 1,340
14 β-Thalassemia 22 10.5 1,500
15 Sickle cell anemia 18 10.7 2,550

From Stein ID: Serum lactate dehydrogenase isoenzymes: Stability, clearance, and diagnostic application in hemolytic
anemia. J Lab Clin Med 1970;76:76–84.

provide an indication of the course of a patient’s of hemolysis. For example, liver disease may affect
hemolytic anemia. LDH, bilirubin, and haptoglobin values, and may also
diminish the marrow’s ability to respond to anemia
with a reticulocytosis. Although hemolysis superim-
Transfusion Requirement posed on liver disease may cause a distinct increase in
An important method for determining the presence of LDH and bilirubin values, these findings may be
hemolysis that is frequently ignored and/or misun- difficult to interpret with certainty. In such situations,
derstood is an evaluation of a patient’s transfusion knowledge of normal transfusion requirements is
requirement. This is particularly significant in patients important, and a significant increase in the absence of
with underlying diseases that affect other indicators bleeding is diagnostic of hemolysis.

5000
Aortic
Mitral
Multiple
Serum Lactic Dehydrogenase Activity (U/L)

r = 0.90
2000

FIGURE 2-10. The correlation between serum lactic


dehydrogenase activity (logarithmic scale) and the T1⁄ 2 of 51Cr- 1000
labeled erythrocytes in 21 patients with ball-valve prostheses. The
unbroken line indicates the regression line; the broken lines show
the 95% confidence interval; and the shaded area shows the
normal values. (From Myhre E, Rasmussen K, Anderson A: Serum 500
lactic dehyrogenase activity in patients with prosthetic heart
valves: A parameter of intravascular hemolysis. Am Heart J
1997;80:463–468.)

200

4 8 12 16 20 24 28 32
T1/2 of 51Cr-Labeled Erythrocytes (days)
The Diagnosis of Hemolytic Anemia 47

32 150

28 120

T1/2 of 51Cr-Labeled Erythrocytes (days)


24

Calculated erythrocyte life span (days)


90

20 60

16
45

12 30

8 20

10
4

100 200 500 1000 2000 5000


Serum Lactic Dehydrogenase (U/L)
FIGURE 2-11. The relationship between serum lactic dehydrogenase, T 1⁄ 2 of Cr-labeled RBCs and
calculated RBC survival in 21 patients with ball-valve prostheses (x); 12 unoperated patients with aortic
valvular disease (° ); and recalculated data from 50 patients with ball-valve prostheses (•). The shaded area
indicates the 95% confidence interval of our regression line. (From Myhre E, Rasmussen K, Andersen A:
Serum lactic dehydrogenase activity in patients with prosthetic heart valves: A parameter of intravascular
hemolysis. Am Heart J 1997;80:463–468.)

NORMAL TRANSFUSION REQUIREMENT expectancy that is one half of normal.51 Accordingly, the
(Table 2-5) daily requirement of transfused RBCs needed to main-
tain a hemoglobin level of 10 g/dL in a 70-kg male who
A 70-kg man has a RBC volume of about 2100 mL is making no RBCs is 14 × 2, or 28 mL/day, or a require-
(30 mL/kg). If, for convenience, a figure of 100 days is ment of 196 mL/week. One unit of RBCs contains about
used as the normal RBC life span, he must produce 180 mL of RBCs, so an average of 1 unit per week will
21 mL of RBCs per day. To maintain a hemoglobin of 10 need to be transfused in order to maintain a hemoglo-
g/dL (about two thirds of normal) requires an RBC pro- bin of 10 g/dL.51 Because transfusion of RBCs sup-
duction of 14 mL/day. Freshly obtained RBCs, taken presses erythropoiesis,51 production of RBCs following
from the circulation of a blood donor, vary in age from transfusion may be significantly reduced. In this case, it
1 to 100 days, and therefore, have an average life should be expected that a patient’s hemoglobin will
return to pretransfusion levels within weeks of a trans-
fusion of 2 or 3 units of RBC; it is incorrect to interpret
TABLE 2-4. APPROXIMATE RBC DESTRUCTION this as an indication of poor survival of transfused
RATE AS PREDICTED FROM THE SERUM LACTIC RBCs.
DEHYDROGENASE LEVELS

Approximate RBC INCREASED TRANSFUSION REQUIREMENT


LDH Destruction Rate AS AN INDICATION OF HEMOLYSIS
(U/L) (× normal)
The knowledge that transfusion of an average of
<200 1 (range, 0.5–1.5)
200–500 2 (range, 1.5–2.5)
1 unit of RBCs per week should be able to maintain a
500–1000 3 (range, 2.0–4.0) reasonable level of hemoglobin in an adult, even if the
>1000 ≥4 patient’s marrow is producing no RBCs, indicates that a
significantly higher transfusion requirement is proof of
Modified from Myhre E, Rasmussen K, Andersen A: Serum lactic
dehydrogenase activity in patients with prosthetic heart valves: A parameter shortened survival of transfused RBCs, provided bleed-
of intravascular hemolysis. Am Heart J 1997;80:463–468. ing can be excluded as a cause. In immune hemolytic
48 Immune Hemolytic Anemias

TABLE 2-5. TRANSFUSION REQUIREMENTS AS AN INDICATION OF HEMOLYSIS

Calculation of normal transfusion requirements for a 70-kg male whose marrow is producing no RBCs

Normal RBC volume = 30 mL/kg = 2100 mL


If the patient’s hemoglobin is 10 g/dL (two thirds of normal), the RBC volume is 1400 mL.
If RBC survival is 100 days, 14 mL of RBCs must be replaced daily to maintain a hemoglobin of 10 g/dL.
Because RBCs obtained from a blood donor are of all ages, average survival of transfused RBCs will be about 50 days.51
Therefore, to maintain a hemoglobin of 10 g/dL, 28 mL would have to be transfused daily, or 196 mL/wk.
Each unit of RBCs contains about 180 mL of RBCs.
Thus, about 1 unit of RBCs per week is a normal transfusion requirement for an adult producing no RBCs.
In the absence of bleeding, a significantly increased transfusion requirement indicates hemolysis, i.e., a short RBC survival time of transfused
RBCs.

anemias and other hemolytic anemias in which hemol- MCHC. Buys and Craven58 reported two such occur-
ysis is due to an extrinsic mechanism, rather than to an rences in patients with C. perfringens infection and com-
intrinsic red cell defect, transfused normal RBCs will mented that other cases have been reported in patients
undergo accelerated destruction. An increased transfu- infected with Leptospira, Toxoplasma, and Plasmodium
sion requirement, therefore, can be an important and organisms, as well as in a patient with AIHA.
definitive indicator of a hemolytic anemia.
Further, a rough estimate of RBC survival can be
HEMOGLOBINEMIA AND HEMOGLOBINURIA
made. For example, if a patient requires 4 units of
RBCs per week to maintain a stable hemoglobin, sur- The cardinal features of intravascular hemolysis are
vival of transfused RBCs must be about one fourth of hemoglobinemia and hemoglobinuria. In normal sub-
normal. If a patient is also producing RBCs, as indi- jects, hemoglobin breakdown occurs mainly in the cells
cated by a reticulocytosis, survival of transfused RBCs of the reticuloendothelial system; therefore, the level of
must be even shorter. free hemoglobin in the plasma is low, ranging from 2 to
5 mg/dL. When hemolysis occurs in the blood, the
Intravascular Hemolysis hemoglobin from the broken down RBCs is liberated
into the plasma, causing an increase in plasma hemo-
Laboratory findings that indicate that hemolysis is globin values to 100 to 200 mg/dL or even more.
primarily intravascular in nature are helpful in that
they suggest that the specific diagnosis is likely to be
one of a limited number of disorders, particularly
those listed in Table 2-6. TABLE 2-6. CAUSES OF HEMOGLOBINURIA
The most remarkable degrees of massive, acute
Acute Hemoglobinuria
intravascular hemolysis in clinical medicine occur as a
result of Clostridium perfringens infection.54-57 The hema- Incompatible blood transfusion
tocrit may drop very rapidly to levels less than 5%.56 Transfusion of damaged blood (overheating or freezing, bacterial
Indeed, a patient reported by Terebelo and coworkers57 contamination, pump-oxygenation)
maintained normal blood pressure, tissue oxygenation, Drugs and chemical agents (immune or nonimmune mechanisms)
Paroxysmal cold hemoglobinuria
and mentation and survived longer than 4 hours after Acute severe warm antibody AIHA
having been found to have a hematocrit of 0 (“total Passenger lymphocyte syndrome
intravascular hemolysis”).56 The peripheral smear dis- Clostridium perfringens infection
closed few intact RBCs. After transfusion of 7 units of Malaria (“blackwater fever”)
RBCs, the hematocrit was 7.2%. Plasma free hemoglo- Bartonellosis, babesiosis, leptospirosis, toxoplasmosis
Peritoneal hemorrhage52
bin was responsible for the preservation of tissue oxy- Severe hypophosphatemia53
genation, intravascular oncotic pressure, and pH. Snake and spider bites
Constitutional symptoms (fever, backache, etc.) Cold agglutinin syndrome*
often accompany acute severe intravascular hemo- March hemoglobinuria
Microangiopathic hemolytic anemia
lysis. Symptoms may begin shortly after the onset of Hypotonic bladder irrigation during prostatic surgery
hemolysis and may be present before the appearance Mistaken intravenous administration of water
of hemoglobinuria.
A distinctive laboratory finding in intravascular Chronic Hemoglobinuria
hemolysis is an elevated mean corpuscular hemoglobin
Paroxysmal nocturnal hemoglobinuria†
concentration (MCHC). This occurs when using an Prosthetic cardiovascular materials
automatic cell counter that calculates the MCHC by
dividing the hemoglobin by the product of the mean *Chronic low-grade intravascular hemolysis is common, with acute
hemoglobinuria resulting from exposure to cold.
corpuscular volume (MCV) and RBCs. The presence of †Characteristically associated with intermittent episodes of grossly evident
plasma free hemoglobin causes the falsely elevated hemoglobinuria.
The Diagnosis of Hemolytic Anemia 49

When the plasma hemoglobin level is markedly hematuria, it is smoky. The microscopic examination
raised, the plasma has a pink or red color, depending of a freshly voided centrifuged specimen will iden-
on the concentration of the hemoglobin. When the rise tify hematuria, as the sediment is seen to contain
is moderate (e.g., from 10 to 40 mg/dL), this color numerous RBCs and the supernatant fluid is clear.
may be lacking, not only because of the relatively low Occasionally, however, the specific gravity of the
concentration but also because other pigments, such urine is so low (<1.007) that the red cells rupture in the
as bilirubin, which gives a yellow color, and met- urine and cause the supernatant to turn red. In such
hemalbumin, which gives a brownish color, may mask cases, the inspection of a carefully collected, cen-
the pink tint. Also, plasma hemoglobin disappears in trifuged specimen of blood, with precautions taken to
2 to 5 hours after the cessation of hemolysis, whereas prevent hemolysis, will distinguish the two condi-
methemalbumin has a half-life of 20 hours.59 tions. In hematuria, the plasma is, of course, of normal
When the level of plasma hemoglobin exceeds the color. Accordingly, red plasma plus red (or dark) urine
renal threshold, hemoglobin appears in the urine, a suggests hemoglobin in plasma and urine; clear
condition termed hemoglobinuria. The urine may be plasma plus red urine suggests hematuria.
pink, red, brown, or almost black.60 It contains two
pigments—oxyhemoglobin and methemoglobin—
CONSEQUENCES OF HEMOGLOBINURIA
that are produced by auto-oxidation of the hemoglo-
bin in the urinary tract when the urine is acidic. In some patients with acute intravascular hemolysis,
Oxyhemoglobin is bright red, whereas methemoglo- the kidneys are damaged, giving rise to acute renal
bin is dark brown; the color of the urine, therefore, failure secondary to acute tubular necrosis. This con-
depends on the concentration (which is related to the dition is characterized clinically by oliguria, which
degree of hemolysis) and the relative proportions of may progress to anuria, uremia, and sometimes,
the two pigments. Oxyhemoglobin predominates in death. Acute tubular necrosis is classically seen fol-
alkaline urine, whereas methemoglobin is predomi- lowing intravascular hemolysis, especially when
nant in acidic urine. Hemoglobinuria usually clears hemolysis is severe. Urinary output should be moni-
within 6 hours of an acute hemolytic episode. It is tored in all cases of acute hemoglobinuria in order to
usually accompanied by albuminuria, which disap- allow early detection of the onset of oliguria.
pears when hemoglobinuria ceases.
The term “blackwater” has been used clinically to
describe the dark color of the urine that occurs in some Hemopexin and Methemalbumin
patients with marked hemoglobinuria. The term has Free heme that is released into the circulation is bound
most commonly been applied to patients with malaria, in a 1:1 ratio to the plasma glycoprotein hemopexin,
in whom hemolysis in association with an acute febrile which is cleared from the plasma with a half-life of
episode has been called “blackwater fever.” 7 to 8 hours.64 A decrease in the hemopexin level
signifies intravascular red cell destruction.
Large quantities of hemoglobin that cannot be
DETECTION OF HEMOGLOBINURIA bound by available haptoglobin or hemopexin are oxi-
Hemoglobin in the urine can be identified by spectro- dized to methemoglobin.59 The heme in this form
scopic examination and by a positive reaction with loses its close affinity for the globin part of the mole-
benzidine and quaiac. Benzidine-positive pigment in cule and is bound by circulating albumin to form
the urine can represent hemoglobin or myoglobin. methemalbumin. It is not rapidly removed from the
Usually, the clinical setting strongly suggests one or circulation (T1/2 = 20 hours), and its presence is con-
the other. Myoglobinuria most commonly occurs after sidered to be a sign of significant intravascular hemol-
crush or other traumatic injury to skeletal muscle, in ysis.59 Methemalbumin may be suspected simply by
intoxicated patients subjected to prolonged muscle visual inspection because it gives the plasma a golden
compression as they lay motionless, and in patients to brown color, depending on its concentration. More
with seizure disorders, although numerous other un- definitive identification may be accomplished by bio-
usual causes have also been reported.61,62 Although chemical tests.
myoglobinuria can mimic hemoglobinuria, the
plasma is less likely to be pink because the small myo- Hemosiderinuria
globin molecule is rapidly cleared into the urine with
Hemoglobinuria results in the deposition in the renal
a half-life of approximately 1 to 3 hours.61,63
tubules of iron-containing granules that are derived
from the breakdown of absorbed hemoglobin and that
DISTINGUISHING HEMOGLOBINURIA are known as hemosiderin. Hemosiderin appears in the
FROM HEMATURIA urine, probably as a result of the desquamation of
tubular cells, and can be demonstrated by staining a
Hemoglobinuria is often confused with hematuria centrifuged urine sediment for iron. Hemosiderinuria is
(RBCs in the urine), especially when the urine is most likely to be seen in patients with chronic intra-
bright red. The urine in hemoglobinuria is clear, but in vascular hemolysis, such as occurs in mechanical
50 Immune Hemolytic Anemias

hemolytic anemias, and it is especially typical of par- span of transfused RBCs decreased from 186 hours to
oxysmal nocturnal hemoglobinuria (PNH), in which 25 hours. They demonstrated that the life span of
hemosiderinuria persists even when hemoglobinuria is transfused RBCs almost normalized following
absent. Indeed, an early term for PNH was “hemolytic splenectomy: a life span of 43 days at postsplenec-
anemia with perpetual hemosiderinuria.”65 tomy day 3 and a life span of 87 days at postsplenec-
Transient hemosiderinuria also occurs in acute tomy day 69.
hemoglobinuria, but it is not found at the onset of a
hemolytic attack, as the pigment has to be absorbed
by the tubular cells of the kidney and reexcreted, a Summary and Comments Concerning
process that takes at least 48 hours.64 The interval the Value of Laboratory Tests to
during which hemosiderinuria may be found follow- Determine the Presence of Hemolysis
ing completion of a transient hemolytic episode has
not been determined, although it has been demon- Hemolytic anemias are relatively uncommon, and
strated that more than 50% of the 59Fe initially present hemolysis is unlikely to be suspected on clinical
in the kidneys still remained 2 weeks after an intra- grounds when a patient presents with a previously
venous injection of 59Fe-labeled hemoglobin.66 undiagnosed anemia. An elevated reticulocyte count
should strongly suggest hemolysis, but reticulocyte
counts may not be part of the initial complete blood
Other Tests count. The presence of hemolysis may unexpectedly
Fehr and Knob67 have evaluated the use of RBC crea- become evident on the basis of laboratory tests ordered
tine as an indicator of the severity of hemolytic for reasons other than for the evaluation of anemia,
disease. In patients with steady-state hemolysis, there such as bilirubin, LDH, or compatibility tests, when a
was a better correlation between RBC survival and blood transfusion is considered to be indicated. In
red cell creatine than there was between RBC survival patients with autoimmune or drug-induced immune
and the reticulocyte count. Only 3 of 21 patients with hemolytic anemias, the laboratory indicators of
hemolytic disease had nomal creatine levels, and hemolysis are usually quite evident and, once con-
these 3 patients had the least reduced 51Cr half-life sidered, the presence of hemolysis is usually easily
values (18.1 to 20 days). The authors suggest that this confirmed by simple tests. These tests include reticu-
simple chemical assay may be used to obtain a useful locyte count, review of the peripheral blood film,
estimation of RBC survival. serum bilirubin (direct and indirect) determinations,
For further evidence of hemolysis, one might assess LDH measurement, and haptoglobin measurement. A
the patient for erythroid hyperplasia of the bone more extensive battery of tests is generally not neces-
marrow or perform RBC survival studies with 51Cr sary, and one should proceed to determining the cause
or 32P-diisopropyl fluorophosphate (DFP)-labeled of the hemolysis, as indicated in the following section.
RBCs.51 Also, measurement of the rate of endogenous Hemoglobinemia and hemoglobinuria are defi-
production of carbon monoxide (CO) may be used to nitive indicators of hemolysis, and their presence
calculate the RBC life span,68-70 and Vreman and should precipitate an urgent evaluation of the patient.
coworkers71 suggest that advances in techniques used However, one should be aware that hemolyzed
for measurement of CO may stimulate the study of plasma may also be caused by in vitro lysis during or
hemolysis. However, at present these tests are not following venipuncture. Thus, the first step in evalua-
generally applied in clinical medicine to establish the tion should be to obtain a repeat blood sample and, in
presence or absence of hemolysis. addition, to inspect the urine visually. Hemoglo-
Burns and coworkers72 have suggested that mea- binemia is generally accompanied by hemoglobin-
surement of RBC adenylate kinase (EAK) is a highly uria, so that if a urine sample has a normal color, in
sensitive and specific test for the diagnosis of hemolytic vitro lysis of the blood sample is to be suspected.
anemia. They studied 25 patients with sickle cell Hemoglobinuria is a much less common clinical
disease, hemolytic transfusion reactions, or thrombotic finding than hematuria, so when the urine is red, the
thrombocytopenic purpura, and compared EAK levels color is often assumed to be the result of the latter.
in those patients with levels found in normal subjects. Centrifugation of the urine specimen will usually
The normal range was 0 to 3.5 units (mean, 0.5); in allow a distinction to be made as to whether the red
patients with hemolysis, the mean level was 62.4 units color is due to RBCs or hemoglobin. In hematuria, the
(range of 0 to 298). The diagnostic sensitivity was 96%, supernatant is clear (although in unusual cases and in
with a specificity and accuracy of 97%. the presence of a low specific gravity of the urine,
Zeiler and coworkers73 used flow cytometry to there may be lysis of RBCs in the urine, causing the
determine the survival of transfused and autologous supernatant to be red). Visual inspection of plasma (or
RBCs in a patient with severe warm antibody AIHA. serum) is helpful because hemoglobinemia and/or
The patient was group A, Rh+, and the authors trans- methemalbuminemia will accompany hemoglobin-
fused group A, Rh– RBCs in order to enable the dis- uria, whereas the plasma will be of normal color in
tinction between autologus and transfused RBCs. The patients with hematuria.
life span of autologous RBCs, measured on two con- Of the easily performed and very valuable tests for
secutive days, was 69 and 64 hours, whereas the life determining the presence of hemolysis, the most
The Diagnosis of Hemolytic Anemia 51

underutilized may be inspection of the peripheral cause of a patient’s hemolytic anemia, it is more prac-
blood film. RBC morphology often strongly suggests tical to emphasize consideration of a limited number
hemolysis and may suggest a specific diagnosis or a of relatively common disorders. The comparatively
limited number of diagnostic possibilities (see page 53). simple classification presented in Table 2-8 is useful
In some complex clinical settings, the usual tests for this purpose.
indicating hemolysis may yield misleading or uninter- As the number of causes of hemolytic anemia is
pretable results. For example, treatment with chemo- large, even when considering the simplified classi-
therapy may supppress the reticulocyte count, and fication, it is evident that it is useful to have a more
liver disease may cause hyperbilirubinemia, elevated restricted list of possibilities in mind before perform-
serum LDH levels, and low serum haptoglobin levels. ing specific diagnostic tests. A tentative or working
In such settings, tests that can be used to determine the diagnosis should be formulated after careful consider-
presence of hemolysis are serial hemoglobin and ation of four important points:
hematocrit determinations (and, when the marrow is
not suppressed, reticulocyte counts). One must keep in 1. A review of the clinical history and physical exam-
mind that there are only two ways to lose RBCs: bleed- ination, especially as they relate to various possible
ing and hemolysis. Accordingly, a marked drop in causes of hemolysis
hemoglobin in a patient who has no evidence of signi- 2. A review of the peripheral blood film
ficant blood loss strongly suggests hemolysis. Because 3. Findings of intravascular hemolysis
blood loss is much more common than hemolysis, 4. The results of a direct antiglobulin test (DAT)
there is a tendency to misinterpret minor amounts of
blood loss, such as that indicated by a weakly positive History and Physical Examination
stool guaiac test, as being an adequate explanation for
a severe anemia or a marked drop in hemoglobin in Because hemolytic anemia is often not suspected on the
patients in whom hemolysis is not anticipated. basis of the initial history and physical examination, it
A definitive indicator of hemolysis that is often mis- is important to review them with emphasis on specific
understood or ignored is the transfusion requirement. points after a diagnosis of hemolysis has been made.
In the absence of bleeding, a significantly increased A history of anemia or of splenectomy in members
transfusion requirement is diagnostic of hemolysis of the family suggests a hereditary hemolytic
(see Table 2-5). Another indicator of hemolysis is the anemia. If present in successive generations, an auto-
presence of a persistent elevation of the reticulocyte somal disorder (such as hereditary spherocytosis) or
count without an increase in the hemoglobin and a condition that is manifested in the heterozygous
hematocrit levels, unless chronic bleeding is present. state (such as the unstable hemoglobins) should be
considered. The most common hereditary RBC
enzyme defects—glucose-6-phosphate dehydroge-
ESTABLISHING A TENTATIVE DIAGNOSIS nase deficiency and pyruvate kinase deficiency—are
OF THE CAUSE OF THE HEMOLYTIC clinically manifested in the sex-linked hemizygous
or rare homozygous state, respectively. In these
ANEMIA instances, the examination of other family members
is of obvious importance. A history of jaundice at
When it has been established that the patient has a birth and long-standing or recurring anemia and/or
hemolytic anemia, the cause of the hemolysis should jaundice may also be obtained in cases of hereditary
be sought next. One should consider the differential hemolytic states.
diagnosis of hemolytic anemias and develop a pre- In contrast, the presence of acquired hemolysis may
liminary diagnosis. Hemolytic anemias have been be indicated by the knowledge of a previously normal
classified in a number of ways. Traditionally, they are blood count or by a history of acute onset of constitu-
divided into intracorpuscular and extracorpuscular tional symptoms, such as fever, malaise, or pain in the
defects or into hereditary and acquired disorders. back, legs, or abdomen.
Efforts have been made to extend these classifications A history of red or dark colored urine suggests the
by incorporating knowledge of the site of the basic presence of hemoglobinuria. The relationship, if any,
defect in reference to the RBC membrane.74,75 Thus, of hemoglobinuria to sleep (PNH), cold (cold agglu-
hemolytic disorders may be caused by intracellular tinin syndrome or PCH), and exertion (march hemo-
abnormalities, defects of the RBC membrane, or extra- globinuria) should also be noted.
cellular abnormalities. Taking into account all the Details relating to drug intake or exposure to chem-
information stated earlier, the authors have developed icals must be obtained, as hemolysis can result from a
a classification system, presented in Table 2-7, which direct toxic effect on red cells, oxidative injury (espe-
considers not only the site of the basic defect, but also cially when an enzyme defect or unstable hemoglobin
the hereditary or acquired nature of many of the rec- is present), or immunologic damage. However, an
ognized causes of hemolytic anemia. accurate history of ingestion of drugs may not always
Although such a classification is comprehensive, it be easy to obtain. Several types of hemolytic anemia
is rather cumbersome, and many of the disorders have been described in alcoholism, particularly when
listed are extremely rare. Thus, when determining the there is associated liver disease.76-80
52 Immune Hemolytic Anemias

TABLE 2-7. CLASSIFICATION OF HEMOLYTIC ANEMIAS

Intracellular Membrane Extracellular


Abnormalities Abnormalities Abnormalities

Hereditary

Enzyme defects Hereditary spherocytosis Lipid abnormalities


Glucose-6-phosphate dehydrogenase, Hereditary stomatocytosis Abetalipoproteinemia (acanthocytosis)
pyruvate kinase deficiency, triosephosphate Rhnull syndrome Lecithin:cholesterol acyltransferase (LCAT)
isomerase deficiency, glucose phosphate Hereditary elliptocytosis deficiency
isomerase deficiency, 2,3-diphosphoglycerate Abnormal cation permeability
mutase deficiency, etc. Hydrocytosis and dessiccytosis
Globin disorders High phosphatidyl choline hemolytic
Defects in globin structure (sickle cell anemia, anemia
hemoglobin S-C disease, hemoglobin S-D Calcium leak with extreme
disease, hemoglobin C disease, etc.; microcytosis
unstable hemoglobins such as hemoglobin Muscular dystrophies
Hammersmith, Bristol, Santa Ana, Madrid) Congenital dyserythropoietic anemias
Defects in globin synthesis (thalassemias)
Heme disorders (porphyrias)
Hepatoerythropoietic porphyria
Congenital erythropoietic porphyria

Acquired

Environmental factors influencing metabolism Paroxysmal nocturnal hemoglobinuria Immune hemolytic anemias
Hypophosphatemia Membrane lipid abnormalities Autoimmune
Wilson’s disease (high serum copper) Liver disease Alloimmune
Uremia Clostridial infection Drug-induced
Severe iron deficiency anemia Anorexia nervosa Drugs (nonimmune)
Vitamin E deficiency Chemicals, venoms
Lead poisoning Mechanical hemolytic anemia
Prosthetic cardiovascular materials
March hemoglobinuria
Microangiopathic hemolytic anemia
Thrombotic thrombocytopenic
purpura
Hemolytic uremic syndrome
Malignant hypertension
Carcinomatosis
Hemangioma
Heat (severe burns)
Infectious agents
Malaria, babesiosis, bartonellosis,
Bacteria: Clostridium perfringins
(welchii),
Mycoplasma pneumoniae
Viral: HIV, CMV, EBV, measles,
mumps, varicella,
Hypersplenism

Hereditary Factor Hereditary and Acquired Acquired Factor

Metabolic defects In conjunction with Oxidant drugs, favism, infections (e.g.,


(Enzymopathies especially glucose viral hepatitis), diabetic ketoacidosis
6-phosphate dehydrogenase deficiency)
Unstable hemoglobins (Zürich, H, Torino, In conjunction with Oxidant drugs, infections
Köln, Shepherds Bush, etc.)

CMV, cytomegalovirus; EBV, Epstein-Barr virus; HIV, human immunodeficiency virus.

Symptoms and signs suggestive of systemic lupus ies. Occasionally, infections are associated with a hemo-
erythematosus, lymphomas, chronic lymphocytic lytic state (see Chapter 3), or they may provoke acute
leukemia, or infections are important because of the hemolysis in subjects with an intrinsic red cell defect.
association of AIHAs with these diseases. Raynaud’s Microangiopathic hemolytic anemia is suggested by
phenomenon and, less frequently, cold urticaria may be the presence of certain underlying disorders, as indi-
present in autoimmune AIHAs caused by cold antibod- cated in Table 2-9.
The Diagnosis of Hemolytic Anemia 53

Basophilic stippling, due to clumping of ribosomes, is


TABLE 2-8. A SIMPLIFIED CLASSIFICATION encountered in many anemias but is prominent in
OF HEMOLYTIC ANEMIAS β-thalassemias, and coarse stippling is seen in lead
poisoning. Hypochromic RBCs associated with a
Hereditary hemolytic anemias
Hereditary spherocytosis
hemolytic state suggest thalassemic disorders, hemo-
Hereditary elliptocytosis globinopathies, lead poisoning, and chronic intravas-
Thalassemias cular hemolysis with urinary loss of iron. Target cells
Hemoglobinopathies (Fig. 2-14) are present in patients with thalassemia or
Enzyme deficiency hemolytic anemias some of the hemoglobinopathies, especially hemoglo-
Acquired hemolytic anemias
Immune hemolytic anemias bins C and E. They may also be found in patients with
Autoimmune hemolytic anemias (AIHAs) obstructive jaundice or hepatitis. Target cell forma-
Warm antibody AIHA tion is more pronounced after splenectomy. Sickle
Cold agglutinin syndrome cells (Fig. 2-15) are seen in patients with sickle cell
Paroxysmal cold hemoglobinuria
Direct antiglobulin test negative AIHA
anemia.
Combined warm and cold AIHA Fragmented RBCs (Fig. 2-16) are typically seen in
Alloimmune hemolytic anemias microangiopathic, mechanical, and some drug-
Hemolytic disease of the fetus and newborn induced hemolytic anemias; irregularly contracted
Hemolytic transfusion reactions red cells of less characteristic appearance are also
Drug-induced immune hemolytic anemia
Drug-induced hemolytic anemias (nonimmunologic mechanisms) present in other hemolytic anemias (see Table 2-9). In
Direct toxic effect hereditary elliptocytosis (or ovalocytosis) (Fig. 2-17),
Idiosyncrasy mechanism 50% to 90% of the RBCs are oval. In normal subjects,
Hemolysis associated with transplantation less than 15% of RBCs are oval, and such cells are also
Hemolytic anemias associated with numerous irregularly
contracted erythrocytes in the blood film
present in a wide variety of anemias, including iron
Mechanical hemolytic anemia deficiency, thalassemia, megaloblastic anemia, and
Microangiopathic hemolytic anemias myelofibrosis. However, the degree of elliptocytosis is
Paroxysmal nocturnal hemoglobinuria most marked in the hereditary form.
Miscellaneous Cold agglutinin syndrome is suggested by the
Infectious agents (uncommon)
Protozoal parasites: malaria finding of gross agglutination in blood films made at
Bacteria: Clostridium perfringens (welchii) room temperature, especially if agglutination is not
present when the same sample of blood is warmed to

The classic physical findings of hemolytic anemia TABLE 2-9. DIFFERENTIAL DIAGNOSIS IN
are pallor, jaundice, and splenomegaly. However, in HEMOLYTIC ANEMIAS ASSOCIATED WITH
milder degrees of hemolysis, these findings are often SPHEROCYTOSIS OR NUMEROUS IRREGULARLY
absent. Leg ulcers or their residual pigmentation, CONTRACTED ERYTHROCYTES
typically over the malleoli, may be present in chronic
hemolytic anemias. Thickening of the skull as a result Spherocytes
Hereditary spherocytosis
of bone marrow expansion may occur in severe hered- Immune hemolytic anemias
itary hemolytic states, and a radiologic examination Alloimmune hemolytic anemia, especially ABO hemolytic
will show thinning of cortical bone with expansion disease of the newborn and hemolytic transfusion reactions
and trabeculation of the medulla, sometimes with the Autoimmune hemolytic anemia, especially warm antibody
typical hair-on-end appearance. AIHA
Drug-induced hemolysis (some cases)
Although splenomegaly occurs in many hemolytic Severe burns
anemias, massive splenomegaly suggests thalassemia Clostridium perfringens (welchii) septicemia
major, lymphoma, myelofibrosis, or chronic Hypophosphatemia
leukemias. Purpura may be seen in hemolytic Fragmented erythrocytes (schistocytes, helmet cells, burr cells)
Chemical or drug-induced hemolytic anemia (some cases)
anemias associated with systemic lupus erythemato- Mechanical hemolytic anemia (prosthetic cardiovascular materials)
sus, thrombotic thrombocytopenic purpura, or Traumatic hemolytic anemia (March hemoglobinuria)
Evans’s syndrome. Microangropathic hemolytic anemia
Hemolytic uremic syndrome
Thrombotic thrombocytopenic purpura
The Peripheral Blood Film Disseminated intravascular coagulation
Malignant hypertension
The peripheral blood film is often very helpful in sug- Disseminated malignancy
gesting a specific diagnosis or a limited number of Less characteristic irregularly contracted erythrocytes
diagnostic possibilities. Spherocytes may be a promi- Severe megaloblastic anemia
Severe iron deficiency
nent feature in several hemolytic anemias, as indi- Thalassemia major
cated in Table 2-9 and in Figures 2-12 and 2-13.
54 Immune Hemolytic Anemias

FIGURE 2-12. Hereditary spherocytosis. Numerous densely staining microspherocytes are present.

FIGURE 2-13. Warm antibody autoimmune hemolytic anemia. Spherocytes are evident, and the degree of
poikilocytosis is generally more marked than in hereditary spherocytosis as a result of the interaction of
antibody and/or complement-sensitized RBCs with macrophages.

37°C prior to making the blood film (Figs. 2-18 and they occur in larger numbers in a rare hemolytic
2-19). Erythroblasts are not infrequently present in the anemia associated with increased red cell sodium
peripheral blood of patients with hemolytic anemia, permeability81 and in alcoholic liver disease.82 Acan-
particularly in infants and children. Stomatocytes are thocytes are characteristically seen in hereditary
present in small numbers in many blood smears, but abetalipoproteinemia, chronic liver disease, and
The Diagnosis of Hemolytic Anemia 55

FIGURE 2-14. Hemoglobin C. Numerous target cells are present. Hemoglobin electrophoresis indicated
that the patient was heterozygous for hemoglobin C.

FIGURE 2-15. Hemoglobin S. Anisocytosis, poikilocytosis, and several sickle cells are illustrated.

certain inherited neurologic disorders, and in associa- examination of the film may indicate the presence of
tion with the inheritance of certain RBC antigen poly- chronic lymphatic leukemia or infectious mono-
morphisms.83 nucleosis. Also, a marked decrease in platelets in a
It is also true that findings in the peripheral blood patient with hemolytic anemia who has numerous
film other than RBC morphology may suggest the fragmented RBCs in the peripheral blood film sug-
cause of a patient’s hemolytic anemia. For example, gests a diagnosis of thrombotic thrombocytopenic
IHA-02(033-060) 11/18/03 4:54 PM Page 56

56 Immune Hemolytic Anemias

FIGURE 2-16. Thrombotic thrombocytopenic purpura. Numerous fragmented RBCs and a marked
decrease in the number of platelets are characteristic features of the disorder. A nucleated RBC is
also present.

FIGURE 2-17. Hereditary elliptocytosis. Occasionally, elliptocytes may be seen in a variety of anemias, but
herediatry elliptocytosis is characterized by the marked degree of elliptocytosus illustrated here. About
15% of patients with hereditary elliptocytosis have hemolytic anemia.
The Diagnosis of Hemolytic Anemia 57

FIGURE 2-18. Cold agglutinin syndrome. The blood film was made at room temperature, and gross
autoagglutination is evident.

FIGURE 2-19. Cold agglutinin syndrome. Same patient as in Figure 2-18, but the blood film was
made strictly at 37°C. Autoagglutination is not present. There is an occasional microspherocyte,
moderate poikilocytosis, and anisocytosis with some macrocytes that are probably reticulocytes.
58 Immune Hemolytic Anemias

purpura or the hemolytic uremic syndrome, and


thrombocytopenia in the presence of spherocytes sug- TABLE 2-10. SCREENING PATIENTS WITH
gests Evans’s syndrome. HEMOLYTIC ANEMIA FOR ACQUIRED IMMUNE
HEMOLYTIC ANEMIA (AIHA)
Intravascular Hemolysis Positive DAT Negative DAT Total
The presence of hemoglobinemia and hemoglobin- AIHA 24 1 25
uria, that is, intravascular hemolysis, is an important No AIHA 5 70 75
clinical finding. Such a finding indicates the likeli- Total 29 71 100
hood that severe hemolysis is taking place and that
Positive value of a positive result = (24/29) × 100 = 83%.
certain disorders should be considered as the most Positive value of a negative result = (70/71) × 100 = 99%.
probable diagnostic possibilities (see page 48 and Modified from Kaplan HS, Garratty G: Predictive value of direct antiglobulin
test results. Diagn Med 1985;8:29–33.
Table 2-6).

The Direct Antiglobulin Test text to describe the details of the use of specific
The direct antiglobulin test (DAT) should be per- confirmatory tests for a wide variety of nonimmune
formed in every patient in whom the presence of hemolytic anemias. However, a few examples are cited
hemolysis has been established. Although some to illustrate the use of the principles advocated in this
exceptions to this rule might be considered, as when chapter.
the diagnosis of a congenital hemolytic anemia is If, in a patient with hemolytic anemia, the history
evident, the DAT is a simple, quick, inexpensive test suggests a hereditary hemolytic anemia, the physical
that yields useful information. examination reveals splenomegaly, the peripheral
A positive result on a DAT in a patient with blood film reveals spherocytes, features of intravascu-
hemolytic anemia does, of course, indicate that the lar hemolysis are absent, and the DAT is negative, a
most likely diagnosis is one of the immune hemolytic tentative diagnosis of hereditary spherocytosis is war-
anemias.84 It should be emphasized that determi- ranted. A family history may reveal a similar diagno-
nation of the presence or absence of hemolysis, as sis in other family members. Osmotic fragility and
described earlier in this chapter, should logically autohemolysis tests will yield characteristic abnor-
precede the performance of the DAT. The purpose of malities, although similar results may be obtained in
the latter test is to indicate the presence or absence of patients who have spherocytosis as a result of
an immune etiology in patients known to have acquired disease. Further specialized testing is rarely
hemolytic anemia (Table 2-10). Indeed, the predictive needed to confirm the diagnosis, although tests may
value of a positive DAT for determining whether or be performed to identify characteristic membrane
not a patient has immune hemolytic anemia is 83% in abnormalities.86,87
a patient with hemolytic anemia but only 1.4% in a If an adult patient with hemolytic anemia mani-
patient without hemolytic anemia.84 festing for the first time has a history of repeated
Although the result of the DAT in a patient with episodes of dark urine and abdominal pain, a blood
hemolytic anemia generally indicates whether or not film that reveals no characteristic abnormalities, labo-
the hemolytic anemia is immunologically mediated, a ratory tests indicating features of intravascular
positive DAT may occur as a coincidental finding in hemolysis, and a negative DAT, the tentative diagno-
patients with hemolytic anemia caused by nonim- sis of PNH may be made. In past years, the diagnosis
mune mechanisms, and some patients with immune was based on the results of Ham’s acidified serum
hemolytic anemias have a negative DAT.85 test, with the sucrose hemolysis test serving as a
Moreover, although a positive DAT is quite unusual screening test. More recently, flow cytometry using
in perfectly healthy persons, positive reactions are antibodies to the glycosylphosphatidylinositol-
obtained in a significant number of hospitalized anchored proteins CD55 and CD59 has been pro-
patients who do not have hemolytic anemia.85 These posed as the standard diagnostic test.88-90 Further
findings emphasize that careful evaluation is required relevant tests include the documentation of hemoglo-
before a precise diagnosis can be made in patients binuria and hemosiderinuria.
who have a hemolytic anemia and a positive DAT test. Another example is that of a patient who has an
The details of such an evaluation are discussed in acquired hemolytic anemia; does not have a history of
Chapters 5 and 6. recent blood transfusion or drug ingestion; has an
acute onset of systemic symptoms, such as fever,
malaise, and aching in the back and legs; a physical
SPECIFIC CONFIRMATORY TESTS examination that reveals splenomegaly; a blood film
revealing spherocytosis; no evidence of hemoglobin-
On the basis of the evaluation indicated thus far, uria; and a strongly positive DAT. An AIHA is the
a provisionaI diagnosis or a limited number of most likely diagnosis, and details of further diagnos-
diagnostic possibilities can usually be established with tic tests that should be performed are described in
reasonable certainty. It is beyond the scope of this later chapters.
The Diagnosis of Hemolytic Anemia 59

R E F E R E N C E S 29. Kaplan M, Hammerman C, Rubaltelli FF, et al: Hemolysis


and bilirubin conjugation in association with UDP-
1. Petz LD. The diagnosis of hemolytic anemia. In: Bell CA (ed): glucuronosyltransferase 1A1 promoter polymorphism.
A Seminar on Laboratory Management of Hemolysis. Hepatology 2002;35:905–911.
Washington, DC: American Association of Blood Banks, 30. Iolascon A, Perrotta S, Coppola B, Carbone R, Miraglia DG:
1979:1–27. Frequency of Gilbert’s syndrome associated with UGTA1
2. Erslev AJ: Compensated hemolytic anemia. Blood (TA)(7) polymorphism in Southern Italy. Haematologica
1995;86:3612–3613. 2000;85:335–336.
3. Finch C: Regulators of iron balance in humans. Blood 31. del Giudice EM, Perrotta S, Nobili B, Specchia C, d’Urzo G,
1994;84:1697–1702. Iolascon A: Coinheritance of Gilbert syndrome increases the
4. Erslev AJ, Silver RK: Compensated hemolytic anemia. Blood risk for developing gallstones in patients with hereditary sphe-
Cells 1975;1:509. rocytosis. Blood 1999;94:2259–2262.
5. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias, 32. White P, Coburn RF, Williams WJ, Goldwein MI, Rother ML,
1st ed. New York: Churchill Livingstone, 1980. Shafer BC: Carbon monoxide production associated with in-
6. Andersen MN, Gabrieli E, Zizzi JA: Chronic hemolysis in effective erythropoiesis. J Clin Invest 1967;46:1986–1998.
patients with ball-valve prostheses. J Thorac Cardiovasc Surg 33. Horne MK, Rosse WF, Flickinger EG, Saltzman HA: “Early-
1965;50:501–510. peak” carbon monoxide production in certain erythropoietic
7. Brus I, Lewis SM: The haptoglobin content of serum in disorders. Blood 1975;45:365–375.
haemolytic anaemia. Br J Haematol 1959;5:348. 34. Berendsohn S, Lowman J, Sundberg D, Watson CJ: Idiopathic
8. Wallerstein RO: Laboratory evaluation of anemia. West J Med dyserythropoietic jaundice. Blood 1964;24:1.
1987;146:443–451. 35. Tisdale WA, Klatskin G, Kinsella ED: The significance of the
9. Todd D: Diagnosis of haemolytic states. Clin Haematol direct-reacting fraction of serum bilirubin in hemolytic jaun-
1975;4:63–81. dice. Am J Med 1959;26:214.
10. Hillman RS, Finch CA: The detection of anemia. In Red Cell 36. Pirofsky B: General Laboratory Observations. Autoimmu-
Manual. Philadelphia: FA Davis, 1969:39–65. nization and the Autoimmune Hemolytic Anemias. Baltimore:
11. Hillman RS, Finch CA: Erythropoiesis: Normal and abnormal. Williams & Wilkins Company, 1969:65.
Semin Hematol 1967;4:327–336. 37. Allgood JW, Chaplin H, Jr: Idiopathic acquired autoimmune
12. Hillman RS: Characteristics of marrow production and reticu- hemolytic anemia. A review of forty-seven cases treated from
locyte maturation in normal man in response to anemia. J Clin 1955 through 1965. Am J Med 1967;43:254–273.
Invest 1969;48:443–453. 38. Schalm L, Weber AP: Jaundice with conjugated bilirubin in
13. Hillman RS, Finch CA: The misused reticulocyte. Br J Haematol hyperhaemolysis. Acta Med Scand 1964;176:549.
1969;17:313–315. 39. Beutler E: Production and destruction of erythrocytes. In
14. Chen Y-H, Wang SC: A simple model for estimating the Beutler E, Lichtman MA, Coller BS, Kipps TJ, Seligsohn U
hemolytic rate in patients with sickle cell anemia. J Lab Clin (eds): Williams Hematology, 6th ed. New York: McGraw-Hill,
Med 1985;105:201–208. 2001:355–368.
15. McCurdy PR, Sherman AS: Irreversibly sickled cells and red 40. Manoharan A: Congenital haptoglobin deficiency. Blood
cell survival in sickle cell anemia: A study with both DF32P and 1997;90:1709a–1709.
51CR. Am J Med 1978;64:253–258. 41. Marchand A, Galen RS, Van Lente F: The predictive value of
16. Milner PF, Charache S: Life span of carbamylated red cells in serum haptoglobin in hemolytic disease. JAMA 1980;
sickle cell anemia. J Clin Invest 1973;52:3161–3171. 243:1909–1911.
17. Rhyner K, Ganzoni A: Erythrokinetics: Evaluation of red cell 42. Shinton NK, Richardson RW, Williams JDF: Diagnostic value of
production by ferrokinetics and reticulocyte counts. Eur J Clin serum haptoglobin. J Clin Pathol 1965;18:114–118.
Invest 1972;2:96–101. 43. Noyes WD, Garby L: Rate of haptoglobin synthesis in normal
18. Peebles DA, Hochberg A, Clarke TD: Analysis of manual retic- man: Determinations by the return to normal levels following
ulocyte counting. Am J Clin Pathol 1981;76:713–717. hemoglobin infusion. Scand J Clin Lab Invest 1967;20:33–38.
19. Greenberg ER, Beck R: The effects of sample size on reticulo- 44. Cullhed I: Serum haptoglobin in cases with Starr-Edwards ball-
cyte counting and stool examination. Arch Pathol Lab Med valve prosthesis. Acta Med Scand 1967;181:321–325.
1984;108:396–398. 45. Langley GR, Owen JA, P Adanyi R: The effect of blood transfu-
20. Brecher G, Schneiderman M: A time-saving device for the sions on serum haptoglobin. Br J Haematol 1962;8:392–399.
counting of reticulocytes. Am J Clin Pathol 1950;20:1079–1083. 46. Fink DJ, Petz LD, Black MB: Serum haptoglobin: A valuable
21. Davis BH, Bigelow NC: Automated reticulocyte analysis: diagnostic aid in suspected hemolytic transfusion reaction.
Clinical practice and associated new parameters. Diagn JAMA 1997;199:615–618.
Hematol 1994;8:617–630. 47. Stein ID: Serum lactate dehydrogenase isoenzymes: Stability,
22. Davis BH, Bigelow NC: Flow cytometric reticulocyte analysis clearance, and diagnostic application in hemolytic anemia. J
and the reticulocyte maturity index. Ann N Y Acad Sci Lab Clin Med 1970;76:76–84.
1993;677:281–292. 48. Myhre E, Rasmussen K, Andersen A: Serum lactic dehydroge-
23. Davis H, Ornvold K, Bigelow N: Flow cytometric reticulocyte nase activity in patients with prosthetic heart valves: A para-
maturity index: A useful laboratory parameter of erythropoi- meter of intravascular hemolysis. Am Heart J 1997;80:463–468.
etic activity in anemia. Cytometry 1995;22:35–39. 49. Walsh JR, Starr A, Ritzmann LW: Intravascular hemolysis in
24. Chang C, Kass L: Clinical significance of immature reticulocyte patients with prosthetic valves and valvular heart disease.
fraction determined by automated reticulocyte counting. Am J Circulation 1969;39(Suppl. 1):135–140.
Clin Pathol 1997;108:69–73. 50. Domen RE: An overview of immune hemolytic anemias. Cleve
25. Moseley RH. Approach to the patient with jaundice. In Humes Clin J Med 1998;65:89–99.
HD (ed): Kelly’s Textbook of Internal Medicine, 45th ed. 51. The transfusion of red cells. In Mollison PL, Engelfriet CP,
Philadelphia: Lippincott Williams & Wilkins, 2000:782–791. Contreras M (eds): Blood Transfusion in Clinical Medicine,
26. Maldonado JE, Kyle RA, Schoenfield LJ: Increased serum con- 10th ed. Oxford: Blackwell Science, 1997:278–314.
jugated bilirubin in hemolytic anemia. Postgrad Med 52. Martin-Nunez G, Fernandez-Galan MA, Lopez-Lopez R,
1974;55:183–190. Gonzalez-Hurtado JA: Peritoneal haemorrhage mimicking
27. Berlin NI, Berk PD: Quantitative aspects of bilirubin metabo- intravascular haemolysis. Br J Haematol 2001;115:238.
lism for hematologists. Blood 1981;57:983–999. 53. Melvin JD, Watts RG: Severe hypophosphatemia: A rare cause
28. Bosma PJ, Chowdhury JR, Bakker C, et al: The genetic basis of intravascular hemolysis. Am J Hematol 2002;69:223–224.
of the reduced expression of bilirubin udp-glucuronosyl- 54. Jimenez M, Sanz C, Alvarez A, Pereira A: Massive intravascu-
transferase 1 in Gilbert’s syndrome. N Engl J Med 1995;333: lar haemolysis in a patient with Clostridium perfringens sepsis.
1171–1175. Vox Sang 2002;82:214.
60 Immune Hemolytic Anemias

55. Garcia-Suarez J, de Miguel D, Krsnik I, Barr-Ali M, Hernanz N, 75. Catlett JP, Petz LD: Hemolytic anemias. In Humes HD (ed):
Burgaleta C: Spontaneous gas gangrene in malignant lym- Kelly’s Textbook of Medicine, 4th ed. Philadelphia: Lippincott
phoma: An underreported complication? Am J Hematol Williams & Wilkins, 2000:1769–1788.
2002;70:145–148. 76. Eichner ER: The hematologic disorders of alcoholism. Am J
56. Chaplin H: Abdominal pain, total intravascular hemolysis, and Med 1973;54:621–630.
death in a 53-year-old woman. Am J Med 1990;88:667–674. 77. Straus DJ: Hematologic aspects of alcoholism. Semin Hematol
57. Terebelo HR, McCue RL, Lenneville MS: Implication of plasma 1973;10:183–194.
free hemoglobin in massive clostridial hemolysis. JAMA 78. Petz LD: Hematologic aspects of liver disease. Curr Opin
1982;248:2028–2029. Gastroenterol 1989;5:372–377.
58. Buys S, Craven C: MCHC in intravascular hemolysis. Am J 79. Morse EE: Mechanisms of hemolysis in liver disease. Ann Clin
Hematol 1990;33:282–283. Lab Sci 1990;20:169–174.
59. Kimber RJ: An approach to the diagnosis of haemolytic 80. Kristensson AA, Wallersted S, Alling C, Cederblad G,
anaemia. Med J Aust 1974;2:532–535. Magnusson B: Haematological findings in chronic alcoholics
60. Berman LB: When the urine is red. JAMA 1977;237:2753–2754. after heavy drinking with special reference to haemolysis. Eur
61. Slater MS, Mullins RJ: Rhabdomyolysis and myoglobinuric J Clin Invest 1986;16:178–183.
renal failure in trauma and surgical patients: A review. J Am 81. Zarkowsky HS, Oski FA, Sha’afi R, Shohet SB, Nathan DG:
Coll Surg 1998;186:693–716. Congenital hemolytic anemia with high sodium, low potas-
62. Hroncich ME, Rudinger AN: Rhabdomyolysis with pneumo- sium red cells. I. Studies of membrane permeability. N Engl J
coccal pneumonia: A report of two cases. Am J Med Med 1968;278:573–581.
1989;86:467–468. 82. Douglass CC, Twomey JJ: Transient stomatocytosis with
63. Hillman RS, Finch CA: Differential Diagnosis of Anemia. In hemolysis: A previously unrecognized complication of alco-
Red Cell Manual. Philadelphia: FA Davis, 1969:112. holism. Ann Intern Med 1970;72:159–164.
64. Sears DA: Disposal of plasma heme in normal man and patients 83. Gallagher PG: Acanthocytosis, stomatocytosis, and related disor-
with intravascular hemolysis. J Clin Invest 1970;49:5–14. ders. In Beutler E, Lichtman MA, Coller BS, Kipps TJ, Seligsohn
65. Marchiafava E: Anemia emolitica con emosiderinuria per- U (eds): Williams Hematology, 6th ed. New York: McGraw-Hill,
petua. Policlinico, Sez med 1928;35:109–120. 2001:519–526.
66. Pimstone NR: Renal degradation of hemoglobin. Semin 84. Kaplan HS, Garratty G: Predictive value of direct antiglobulin
Hematol 1972;9:31–42. test results. Diagn Med 1985;8:29–33.
67. Fehr J, Knob M: Comparison of red cell creatine level and retic- 85. Garratty G: The significance of IgG on the red cell surface.
ulocyte count in appraising the severity of hemolytic processes. Transfus Med Reviews 1987;1:47–57.
Blood 1979;53:966–976. 86. Gallagher PG, Forget BG: Hereditary spherocytosis, elliptocy-
68. Coburn RF, Williams WJ, Kahn SB: Endogenous carbon monox- tosis, and related disorders. In Beutler E, Lichtman MA, Coller
ide production in patients with hemolytic anemia. J Clin Invest BS, Kipps TJ, Seligsohn U (eds): Williams Hematology. New
1966;45:460–468. York: McGraw-Hill, 2001:503–518.
69. Engel RR, Rodkey FL, Krill CE: Carboxyhemoglobin levels as 87. Dacie J: The Haemolytic Anaemias, 3rd ed. vol. 1, The
an index of hemolysis. Pediatrics 1971;47:723–730. Hereditary Haemolytic Anaemias. New York: Churchill
70. Landaw SA, Winchell HS: Endogenous production of 14CO: A Livingstone, 1985.
method for calculation of RBC lifespan in vivo. Blood 88. Hall SE, Rosse WF: The use of monoclonal antibodies and flow
1970;36:642–656. cytometry in the diagnosis of paroxysmal nocturnal hemoglo-
71. Vreman HJ, Mahoney JJ, Stevenson DK: Carbon monoxide and binuria. Blood 1996;87:5332–5340.
carboxyhemoglobin. Adv Pediatr, 1995;42:303–334. 89. van der Schoot CE, Huizinga TW, van ‘t Veer-Korthof ET,
72. Burns ER, Kale A, Murthy VV: Diagnosis of the hemolytic state Wijmans R, Pinkster J, von dem Borne AE: Deficiency of glyco-
using serum levels of erythrocyte adenylate kinase. Am J syl-phosphatidylinositol-linked membrane glycoproteins of
Hematol 2000;64:180–183. leukocytes in paroxysmal nocturnal hemoglobinuria, descrip-
73. Zeiler T, Muller JT, Hasse C, Kullmer J, Kretschmer V: Flow tion of a new diagnostic cytofluorometric assay. Blood
cytometric determination of RBC survival in autoimmune 1990;76:1853–1859.
hemolytic anemia. Transfusion 2001;41:493–498. 90. Beutler E: Paroxysmal nocturnal hemoglobinuria. In Beutler E,
74. Ballas SK: Disorders of the red cell membrane: A reclassi- Lichtman MA, Coller BS, Kipps TJ, Seligsohn U (eds): Williams
fication of hemolytic anemias. Am J Med Sci 1978;276:4–22. Hematology, 6th ed. New York: McGraw-Hill, 2001:419–424.
C H A P T E R 3

Classification and Clinical


Characteristics of
Autoimmune Hemolytic
Anemias

CLASSIFICATION We refer to AIHA as idiopathic if it is unassociated


with any demonstrable underlying disease. If the
In this chapter we present a AIHA is associated with an additional disorder and
classification of immune he- there is reason to suspect that the association is not
molytic anemias and review merely fortuitous, we refer to the AIHA as secondary.
the clinical manifestations of AIHA associated with infections is generally transient
each of the autoimmune he- and remits after resolution of the infectious disease. In
molytic anemias (AIHAs). patients with an associated lymphoproliferative dis-
Numerous classifications of order, the course of the AIHA is less predictable. A
immune hemolytic anemias significant distinction between idiopathic and second-
have been proposed.1 The purpose of a classification ary cases of CAS is that the IgM cold agglutinin in
of any group of diseases should be to divide that patients with secondary disease associated with infec-
group into clinically distinctive categories. The rather tions is a polyclonal protein, in contrast to the mono-
simple classification listed in Table 3-1 serves this clonal antibody that is characteristically found in
purpose well because the clinical manifestations, patients with idiopathic disease or with an underlying
prognosis, and therapy differ among the diagnostic lymphoproliferative disorder.
groups listed. Table 3-2 outlines some of these Some patients have both warm and cold autoanti-
distinctions. bodies in their sera, and the serologic characteristics of
The following are some comments about the these antibodies may satisfy the criteria for both warm
classification listed in Table 3-1. The majority of cases antibody AIHA and CAS. We refer to these cases as
of AIHA are mediated by warm-reactive autoantibodies, instances of “combined cold and warm AIHA,” or
that is, antibodies that display optimal reactivity with “mixed AIHA.”
human red blood cells (RBCs) at 37°C and that are Paroxysmal cold hemoglobinuria (PCH) is distinct
usually of the immunoglobulin G (IgG) class. In con- from cold agglutinin disease. The causative anti-
trast, cold agglutinin syndrome (CAS) is generally body is an IgG immunoglobulin with specificity that
caused by IgM autoantibodies that exhibit maximal differs from that found in cold agglutinin disease.
reactivity at 4°C. The distinction between cold and The antibody is best detected in vitro by its ability to
warm AIHAs (WAIHAs) is important, because the cause hemolysis of normal RBCs in a two-step
prognosis and management differ significantly. procedure, which requires incubation in the cold

61
62 Immune Hemolytic Anemias

TABLE 3-1. CLASSIFICATION OF IMMUNE TABLE 3-2. SOME CHARACTERISTIC FEATURES


HEMOLYTIC ANEMIAS OF AUTOIMMUNE AND DRUG-INDUCED IMMUNE
HEMOLYTIC ANEMIAS
Autoimmune hemolytic anemias (AIHAs)
Warm antibody AIHA Warm Antibody Autoimmune Hemolytic Anemia
Idiopathic
Secondary (e.g., chronic lymphocytic leukemia, lymphomas, Clinical manifestations: Variable, usually symptoms of anemia,
systemic lupus erythematosus) occasionally acute hemolytic syndrome
Cold agglutinin syndrome Prognosis: Fair, with significant mortality
Idiopathic Most effective therapies: Steroids, splenectomy, immunosuppressive
Secondary drugs
Nonmalignant disorders (e.g., mycoplasma pneumoniae
infection, infectious mononucleosis, other virus infections) Cold Agglutinin Syndrome
Malignant disorders (e.g., lymphoproliferative disorders)
Paroxysmal cold hemoglobinuria Clinical manifestations: Moderate chronic hemolytic anemia in
Idiopathic middle-aged or elderly person, often with signs and symptoms
Secondary exacerbated by cold
Viral syndromes Prognosis: Good, usually a chronic and quite stable anemia
Syphilis Most effective therapies: Avoidance of cold exposure,
Combined cold and warm AIHA (“mixed AIHA”) immunosuppressive drugs
Atypical AIHA
AIHA with a negative direct antiglobulin test Paroxysmal Cold Hemoglobinuria
Warm antibody AIHA caused by IgM or IgA autoantibodies
Drug-induced immune hemolytic anemia Clinical manifestations: Acute hemolytic anemia, often with
Drug-related antibody identifiable hemoglobinuria, particularly in a child with history of recent viral
Drug-induced AIHA or viral-like illness
Alloantibody-induced immune hemolytic anemia Prognosis: Excellent after initial stormy course
Hemolytic transfusion reactions Therapy: Not well defined; steroids empirically and transfusions if
Hemolytic disease of the fetus and newborn required

Drug-Induced Immune Hemolytic Anemia


followed by incubation at 37°C in the presence of Clinical manifestations: Variable, most commonly subacute in onset,
complement. but occasionally acute hemolytic syndrome
The ingestion of some drugs causes hemolytic Prognosis: Excellent
anemia, in which the causative antibody can be shown Therapy: Stop drug; occasionally a short course of steroids
to have specificity for the drug or its metabolites. We empirically
do not consider these cases of drug-induced immune
hemolytic anemia to be autoimmune disorders
because the antibody does not have specificity for lation. Similarly, data obtained from a hospital in
autoantigens. In other quite remarkable cases, the Odense, Denmark, that received all medical cases
ingestion of a drug causes the development of RBC from a population of 230,000 indicated an annual
autoantibodies, that is, the antibody in the patient’s incidence of about 1:75,000 population.2 A study
serum and in an eluate from the patient’s red cells during a 5-year period in one of Sweden’s health care
reacts with red cells similarly to autoantibodies in regions indicated an incidence of 2.6:100,000 persons
idiopathic warm antibody AIHA, and no relationship per year.3 Also, Sokol and coworkers4 reported a
between the drug and the antibody can be demon- series of 1694 patients with RBC autoantibodies and
strated in vitro. Such cases are appropriately termed stated that the incidence of AIHA was about 1:41,000.
drug-induced AIHA. These authors included patients who had hemolysis
The alloantibody-induced hemolytic anemias are in the absence of anemia and commented that the rel-
reviewed in Chapters 13 and 14. atively high incidence they observed was probably
due to the inclusion of patients with mild and com-
pensated hemolysis.
CLINICAL CHARACTERISTICS OF WAIHA is by far the most common type of AIHA
AUTOIMMUNE HEMOLYTIC ANEMIAS and represented 70.3% of the 347 patients in our
series1 (Table 3-3). Dacie and Worlledge5 reported
284 patients with AIHA, and 70% of the cases were
Warm Antibody AIHA WAIHA. Engelfriet and coworkers6 reported that 83%
INCIDENCE of their 2390 cases of AIHA were caused by warm
autoantibodies and, in another 0.4%, both warm and
In Portland, Oregon, Pirofsky2 reported findings in cold autoantibodies were present. Sokol and cowork-
patients who were referred from a population of ers4 classified 60% of their patients as having warm
2.3 million in the Pacific Northwest of the United autoantibodies (excluding drug-induced immune
States over an 8-year period. He concluded that the hemolytic anemia) and another 7.5% as having both
minimum annual incidence of AIHA is 1:80,000 popu- warm and cold autoantibodies.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 63

12
TABLE 3-3. INCIDENCE OF VARIOUS KINDS Warm autoimmune
OF ACQUIRED IMMUNE HEMOLYTIC ANEMIAS hemolysis
IN OUR SERIES OF 347 PATIENTS 10 Cold autoimmune

Incidence per 100,000 of population


hemolysis +
No. of paroxysmal cold
Patients Percent of Total 8 hemoglobinuria
Mixed autoimmune
Warm AIHA 244* 70.3
Cold agglutinin syndrome 54 15.6 hemolysis
Paroxysmal cold hemoglobinuria 6 1.7 6
Drug-induced IHA 43 12.4

IHA, immune hemolytic anemia; AIHA, autoimmune hemalytic anemia;


4
WAIHA, warm AIHA.
* Includes 30 patients with WAIHA induced by α-methyldopa.

AGE DISTRIBUTION 2

Subjects of all ages are affected, from infants in the


first few months of life to elderly people.7 Pirofsky8 0
reported ages of onset of 1 month to 87 years. Of these 0–
9
–1
9
–2
9
–3
9 9
– 4 0 –5
9 9
– 6 0 –7
9 +
80
patients, 73% were over 40 years of age, and the peak 10 20 30 40 5 60 7
incidence appeared between the ages of 60 and 70. Age (years)
Sokol and coworkers4 reported that, except for a FIGURE 3-1. Incidence of acquired immune hemolytic anemia per
childhood peak of paroxysmal cold hemoglobinuna 100,000 of population at risk, related to age at presentation. The early
(PCH), there is a general increase in incidence through- childhood peak of cold autoimmune hemolysis (arrow) is largely due to
out life, with a dramatic rise occurring after the age of patients with paroxysmal cold hemoglobinuria. (From Sokol RJ, Booker
DJ, Stamps R: The pathology of autoimmune haemolytic anaemia.
50 (Fig. 3-1). Böttiger and Westerholm3 also found a J Clin Pathol 1992;45:1047–1052.)
marked increase in incidence after the age of 50.
As might be anticipated, the underlying disease
process has a marked effect on the age distribution. In In secondary WAIHA, the percentages are more
Allgood and Chaplin’s9 report of idiopathic cases, the varied, perhaps depending on the incidence of under-
peak incidence was in the fourth to seventh decades, lying diseases seen in referral centers. For example,
with the mean age of onset of 48.6 years. However, Pirofsky2 reported that 80% of cases associated with
patients with secondary warm antibody AIHA associ- SLE occurred in females, whereas a male predomi-
ated with chronic lymphatic leukemia (CLL) or nance was found when patients with lymphoprolifer-
systemic lupus erythematosus (SLE) have the age dis- ative disorders were analyzed.
tribution of the underlying disease. This finding
emphasizes that clinical features of AIHA occurring in CLINICAL MANIFESTATIONS (Table 3-4)
association with other diseases are frequently domi-
nated by underlying pathologic states. WAIHA is an extremely variable disorder, and almost
AIHA in the young is not a rarity, and a large body every grade of severity may be encountered. In some
of literature is available that describes such patients. patients, the onset is slow and insidious over a period
Although clinical and laboratory findings in children of months, with the ultimate emergence of sympto-
are quite similar to those in adults, there are some matic anemia. In other patients, the onset is sudden,
distinctive features, which are emphasized in with fever, abdominal or back pain, malaise, and
Chapter 9. manifestations of rapidly increasing anemia. In severe
anemia, neurologic manifestations may occur that
SEX DISTRIBUTION progress from obtundation to coma and death. In
children, the disease frequently appears as an acute and
Most observers have reported a somewhat higher sometimes fulminating disorder.11,12 Although such
incidence in females than in males. In idiopathic cases acute manifestations are serious and present difficult
of WAIHA, Allgood and Chaplin9 reported that 60% problems in management, they may be of short dura-
of 47 patients were female; Dausset and Colombani10 tion, with complete recovery within a few weeks.11-13
reported that 61% of 93 patients were female; Symptoms that relate directly to the presence of
Pirofsky2 indicates that 64% of 44 patients were anemia and that resolve on improvement in the level
female; Dacie7 reported 58% of 108 patients were of hemoglobin include dizziness, palpitations, and
female; Dacie and Worlledge5 reported that 59% of dyspnea on exertion. In more severely affected
111 patients were female; and, in the series reported patients, the intensity of the anemia often leads to
by Böttiger and Westerholm,3 women predominated serious dyspnea and incapacity.7 Angina, edema, and
in all age groups with the exception of the youngest frank congestive heart failure occur in a small per-
(0 to 14 years), in which the sex distribution was even. centage of patients. At the other end of the scale, the
64 Immune Hemolytic Anemias

pedic and of little or no value. However, considering


TABLE 3-4. WARM ANTIBODY AUTOIMMUNE only those symptoms that may relate specifically to
HEMOLYTIC ANEMIA—CHARACTERISTIC CLINICAL the hemolytic state, weakness is the most common
MANIFESTATIONS complaint; it occurred in 87% of Pirofsky’s series.2
Other common symptoms are fever and jaundice,
Symptoms
although neither of these symptoms occurs in a major-
Symptoms of anemia ity of patients.
Fatigue
Weakness
Dyspnea on exertion
Fever PHYSICAL SIGNS
Abdominal or back pain
Malaise The diagnosis of anemia on the basis of physical signs
Dark urine is remarkably difficult, and the classic observation of
pallor is quite unreliable. Jaundice is more commonly
Physical Signs observed; it occurred as a presenting sign in 39% of
Jaundice the patients in Pirofsky’s series.2
Splenomegaly Splenomegaly was present in 57% of patients with
Hepatomegaly idiopathic WAIHA reported by Allgood and Chaplin,9
Lymphadenopathy and in just over 50% of the patients with either
Venous thromboembolism
idiopathic or secondary AIHA reported by Pirofsky.2
Laboratory Findings Dameshek and Schwartz15 reviewed the data in
40 recorded cases of AIHA and reported that the
Anemia spleen could be palpated in 28 (70%). The organ is
Abnormal red blood cell morphology generally firm, nontender, and only slightly to moder-
Spherocytosis, anisocytosis, poikilocytosis, polychromatophilia,
autoagglutination
ately enlarged. It is unusual for an enlarged spleen to
Reticulocytosis (reticulocytopenia in some patients) reach the umbilicus and, on the whole, the degree of
Thrombocytopenia (“Evans’s syndrome”) splenomegaly is less than that found in hereditary
Leukocytosis (leukopenia in some patients) spherocytosis.
Urine may contain bile pigments (and/or hemoglobin in patients with Hepatomegaly has been reported in about one third8
brisk hemolysis)
Erythroid hyperplasia in the bone marrow to two thirds16 of the patients with idiopathic AIHA,
and the organ is usually firm and nontender. Liver
enlargement is generally moderate, and massive
hepatomegaly is rare. However, liver function may be
patient may be free from symptoms and not be seriously affected in very seriously ill patients and, in
significantly anemic, despite definitive evidence of fatal cases, has usually been described as enlarged.17
hemolysis. Shirey and coworkers,18 for instance, described a female
A history of dark urine may be elicited; this condi- patient aged 22 who had recurrent episodes of intravac-
tion may result from the presence of bile pigments or ular hemolysis. Her liver progressively enlarged and its
hemoglobinuria (see Chapter 2). Hemolytic jaundice function deteriorated. Eventually, she died in hepatic
is classically regarded as acholuric but, in seriously ill coma. The liver failure was attributed to autoagglutina-
patients, significant amounts of conjugated bilirubin tion within the liver sinuses. The antibody responsible
may circulate in the plasma, and bile pigment may was a warm-reacting IgM (see Chapter 5).
appear in the urine.7 Furthermore, urobilinogen fre- The incidence of lymphadenopathy varies in
quently darkens the color of the urine, especially on reported series, depending, at least in part, on the rel-
standing. Hemoglobinuria may cause the urine to be ative proportion of idiopathic and secondary cases. In
pink, red, brown, or almost black, depending on the idiopathic AIHA, the lymph nodes are usually not
concentration and relative proportions of oxyhemo- enlarged.7 However, Pirofsky2 reported that lym-
globin, which is bright red, and methemoglobin, phadenopathy was present in 23% of the patients with
which is dark brown. The final color of the urine is idiopathic WAIHA and 37% of the patients with sec-
determined by the concentration of hemoglobin, the ondary AIHA, a majority of whom had lymphoprolif-
pH of the urine, and duration of contact between erative disorders. Indeed, enlargement of the various
hemoglobin and urine.14 Hemoglobinuria is quite reticuloendothelial structures was the most consistent
unusual in adults, but it is a prominent finding in the abnormality on physical examination, and only 26%
most seriously ill patients. It is much more common in of all patients manifested no enlargement of the
children. spleen, liver, or lymph nodes.
In patients with secondary AIHA, the associated Thromboembolism. Venous thromboembolism (VTE)
pathologic state may cause the most prominent of the has long been recognized as a cause of morbidity and
patient’s symptoms and obscure the symptoms of mortality in AIHA, and an illustrative case reported by
the hemolytic anemia. Considering the diversity of Swisher19 is illustrated in Figure 3-2. VTE have been
underlying diseases associated with AIHA, a com- reported to be responsible for or contributed to the
plete list of presenting symptoms would be encyclo- death of 3% to 10% of patients with AIHA.9,10,20
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 65

Pregnant
Delivery Pregnant Thrombophlebitis
? Pulmonary
Splenectomy embolus Delivery Delivery

(mg. per day)


Corticoid
200

100

40
Reticulocytes (percent)

Hematocrit
Hematocrit (percent)

30

20

10
Reticulocytes

June Sept. Dec. 1953 1954 1955 1956 1957 1958 1959
1952
FIGURE 3-2. Clinical course of idiopathic acquired immune hemolytic anemia in a 25-year-old woman through
repeated pregnancies. (From Swisher SN: Acquired hemolytic disease. Postgrad Med 1966;40:378–386.)

Pullarkat and coworkers21 prospectively studied 30 strong risk factors for thrombosis regardless of the site
patients with AIHA and found that 8 (27%) had a and type of thrombosis. Their detection in patients with
documented VTE, all of which were confirmed by SLE and/or previous thrombosis is therefore justified.
objective imaging studies. In four patients the VTE Analysis of anticardiolipin antibodies was more
occurred with the initial hemolytic crisis, and in three complex because of the effects of isotypes and titer, but
patients thrombotic events occurred with relapse of it appeared that medium or high titer IgG anticardi-
their AIHA. One patient developed a deep vein throm- olipin antibodies helps identify patients at risk for
bosis and pulmonary embolus 3 weeks after laparo- thrombosis. However, these researchers pointed out
scopic splenectomy. The authors determined that 19 of that there is an urgent need to harmonize investiga-
the 30 patients (63%) with AIHA had antiphospholipid tional methods.
(Apl) antibodies, 9 (31%) were positive for the lupus Thrombosis in hemolytic anemia has largely been
anticoagulant (LAC), and 17 (57%) had anticardiolipin attributed to disruption and loss of the RBC mem-
(aCL) antibodies. Seven patients had both LAC and brane, resulting in surface exposure of the negatively
aCL. In the patients with aCL, five were positive for charged phosphatidylserine (PS). Exposed RBC PS is
IgG and 12 had IgM. None had detectable IgA aCL. thought to promote thrombosis by providing a surface
Venous thromboembolic events were observed in five for formation of the tenase and prothrombinase
of the nine patients (63%) positive for LAC. This was complexes.21 In addition to AIHA, VTE complications
statistically significant with a calculated relative risk of have also been reported in patients with inherited
7.5 (95% confidence interval [CI], 1.25 to 45.2, hemolytic anemias, including hereditary spherocyto-
P = 0.03) compared with the patients with VTE and no sis and thalassemia major.23,24 However, the incidence
detectable LAC. No significant association was found of thrombosis appears to be much lower in inherited
between the presence of aCL and the development of hemolytic anemia.
VTE. Further data have been provided by Kokori Additional reports of thrombotic events in patients
and coworkers,22 who found that the patients with SLE with AIHA have been reported including upper
who had AIHA were more likely to have thrombotic extremity arterial thrombosis25,26; superior sagittal
episodes than were SLE patients without AIHA. Galli sinus thrombosis associated with Evans’s syndrome27;
and coworkers22a reviewed the medical literature from mesenteric and portal vein thrombosis28; in patients
1988 to 2000 to formally establish the risk of lupus anti- with cold and WAIHA,29,30 of whom one had multiple
coagulants and anticardiolipin antibodies for arterial pulmonary emboli and extensive deep venous throm-
and venous thrombosis. They calculated the odds ratio bosis of the iliofemoral vessels leading to frank
with 95% confidence interval of lupus anticoagulants gangrene requiring an above-knee amputation30; in a
and/or anticardiolipin antibodies for thrombosis in patient with cold agglutinin disease31; in association
4184 patients and 3151 controls. They concluded that with human immunodeficiency virus (HIV) infection32;
lupus anticoagulants have an odds ratio for thrombosis and after blood transfusion in patients with HIV
5 to 16 times higher than controls and that they are infection.32,33
66 Immune Hemolytic Anemias

Although prophylactic anticoagulation is not indi- with WAIHA. They are present in large numbers when
cated for patients with LAC in the absence of prior hemolysis is severe and if the reticulocyte count is
thrombosis,34 Pullarkat and coworkers21 suggest markedly raised. The largest numbers are seen in
prophylactic anticoagulation for all patients with patients in whom increased hemolysis and anemia
AIHA who are positive for LAC, at least during have persisted after splenectomy. Then the erythroblast
episodes of hemolysis. count may even exceed the total leukocyte count.36
Leukocytes. The white blood cell (WBC) count is
THE BLOOD PICTURE normal in more than half of the patients with idio-
pathic AIHA2,9 but, of course, it varies greatly among
Anemia. The degree of anemia is variable, but it is patients with secondary AIHA, depending on the
not infrequently severe. Allgood and Chaplin9 reported underlying disease. Even in patients with idiopathic
that the initial hemoglobin concentration in 21 of 47 disease, a range of values extending from 1400 to
patients was less than 7 g/dL. Although only a moder- 37,000/μL was obtained in Pirofsky’s series.2 Leuko-
ate anemia may be present on admission, a meticulous cyte counts of less than 2000/μL were observed in 6 of
follow-up is required, particularly when the patient is 38 patients. The latter results occurred in a pattern of
first observed. Progression of the severity of the anemia peripheral pancytopenia with hypoplastic or normal
frequently occurs before therapy becomes effective, bone marrows and may be a manifestation of an
and the fall in hemoglobin and hematocrit may be immune pancytopenia.
rapid, justifying the terms “acute hemolytic anemia” or In acute hemolytic episodes, however, leukocytosis
“hemolytic crisis.” Indeed, Pirofsky8 emphasizes that is frequent, with counts usually ranging from 15,000
among 213 patients, the lowest hematocrit ranged from to 25,000/μL. This finding is chiefly the result of an
7.5% to 41.5%, but with a medium value of only 19%. A increase in neutrophils with occasional metamyelo-
hematocrit of 15% or less was observed in 49 patients! cytes and myelocytes as part of a leukemoid reaction.
Similarly, Crosby and Rappaport35 reported hemo- Counts considerably in excess of the preceding
globin values of 5 g/dL or less in 15 of 34 patients. figures have been reported, especially in children. The
Characteristically, the anemia is macrocytic rather than highest WBC count in Pirofsky’s series2 was in a 21⁄2-
normocytic, with the macrocytosis being associated year-old infant in whom the count was 37,000/μL,
with a raised reticulocyte count. chiefly lymphocytes. Twenty-two of 46 patients report-
Blood Films. Anisocytosis is usually a marked ed by Habibi and coworkers13 had leukocytosis with
feature; this is brought about by the presence of micros- total WBC counts as high as 40,000/μL and with promy-
pherocytes with reduced RBC diameter and macro- clocytes and myelocytes accounting for up to 20% of the
cytes, which are the reticulocytes. Poikilocytes are total.
generally not conspicuous in AIHA. However, in some Liesveld and coworkers44 reviewed the initial
patients small numbers of pear-shaped RBCs may be leukocyte counts of 108 patients with various types of
present. Polychromasia is often obvious, reflecting the autoimmune hemolysis. The counts ranged from 1.5
height of the reticulocyte count.36 to 137 × 109/L; the mean was 10.6 × 109/L, and the
Spherocytosis is often easily recognized in the blood median was 9.0 × 109/L. Forty percent of the patients
film of patients with WAIHA. However, they are not had initial counts exceeding 11 × 109/L. (Three
always present, at any rate to a marked degree, even patients with chronic lymphocytic leukemia were
if the patient is actively hemolyzing.36 excluded from the analysis.)
Autoagglutination in vitro is a phenomenon occa- A reaction to “stress” plus stimulation by products
sionally observed, particularly in more severely of intravascular hemolysis are probably important
affected patients.37 The agglutination typically con- factors in producing the leukocytosis, and similar
sists of rather fine clumps of RBCs that may be visible rises have been reported in instances of hemolysis that
to the naked eye on close observation and, in contrast are not caused by an immune mechanism.36
to those caused by cold agglutinins, they do not Platelets. Platelet counts are usually normal in idio-
change after incubation at 37°C (see Chapter 5). pathic WAIHA. A minority of patients have thrombo-
The presence in the peripheral blood of monocytes cytosis that, on occasion, reaches impressive heights.
(or rarely neutrophils) that have phagocytosed RBCs Allgood and Chaplin9 reported that 24% of their
is a pointer to an autoimmune hemolytic anemia. patients had thrombocytosis at the time of presenta-
Erythrophagocytosis is most easily seen in films made tion, with the highest value recorded being
from the buffy coat of peripheral blood. Sometimes, 1,900,000/μL. Pirofsky8 reported that 17% of his
however, so many erythrophages are present that they patients with AIHA in the absence of malignant lym-
can be found without too much difficulty in films of phoid disease had thrombocytosis, with the highest
whole uncentrifuged blood. Numerous case reports in value being greater than 1,000,000/μL. In the series of
the older medical literature emphasized the presence of Liesveld and coworkers,44 16 of 85 patients had
erythrophagocytosis.7,38-43 One wonders if inspection thrombocytosis (platelet count >400,000/μL). Thirteen
of the blood film has become a less common skill of these patients had warm autoantibodies, two had
among physicians in modern times (see page 76). secondary cold-antibody AIHA, and one patient had
Erythroblasts, mainly polychromatic normoblasts, both warm and cold autoantibodies. Heisel and
are often present in the peripheral blood of patients Ortega45 recorded a mean platelet count at diagnosis
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 67

of 421,000/μL (range, 191,000 to 583,000/μL) in 9 chil- been reported in which AIHA supervened in patients
dren who had acute AIHA and a mean count of who had undergone splenectomy several years earlier
195,000/μL (range, 13,000 to 795,000/μL) in 16 chil- for thrombocytopenic purpura.7 Waugh55 described a
dren who had chronic AIHA (10 had counts patient who died of fulminating hemolytic anemia
<150,000/μL at diagnosis). In contrast, Habibi and and who had undergone splenectomy for chronic
coworkers13 did not mention the occurrence of throm- thrombocytopenic purpura 4 years previously.
bocytosis in their review of 80 children with AIHA, Allgood and Chaplin9 mentioned one patient who
and Crosby and Rappaport,35 who tabulated the developed AIHA 25 years after splenectomy for
platelet counts of 29 patients, recorded only two thrombocytopenic purpura, and they emphasized
counts above 400,000/μL. that the two disorders may be entirely unrelated in
Evans’s Syndrome. Thrombocytopenia occurs in respect to time of onset and response to steroid
some patients in all large series of WAIHA. In 1949, therapy and/or splenectomy.
Evans and Duane46 called attention to this association, Pui and coworkers56 concluded that the combina-
and the simultaneous occurrence of thrombocytope- tion of ITP and AIHA is rare in childhood. Among 164
nia and AIHA is now well recognized and is fre- instances of ITP and 15 instances of AIHA, 11 patients
quently referred to as “Evans’s syndrome.” Dausset were found to have this combination. Three were
and Colombani,10 in a review of 83 patients with idio- found to have SLE, one had aplastic anemia, and
pathic WAIHA, encountered thrombocytopenic seven had Evans’s syndrome. Neutropenia, at times
purpura in 11 (13.2%). Allgood and Chaplin9 reported associated with bacterial infections, occurred in four
thrombocytopenia in 3 of 47 patients at the time of of the latter patients. Unlike most cases of ITP or
diagnosis of the hemolytic anemia; 3 others either had AIHA in childhood, the clinical course of Evans’s syn-
a history of thrombocytopenia, or thrombocytopenia drome was usually chronic and relapsing. Treatment
developed at some time after the diagnosis of AIHA including corticosteroids, splenectomy, and immuno-
was made. Subsequently, many other reports of suppressive agents was generally unsatisfactory. In
patients having both AIHA and severe thrombocy- view of the frequent presence of antibodies directed at
topenia have been described,47-52 and it is now RBCs, platelets, neutrophils, and lymphocytes, the
realized that low platelet counts are found quite authors suggested that immunopancytopenia may be a
commonly in AIHA and that clinically obvious better term for this condition (see later).
purpura is not infrequent.7 Mathew and coworkers51 conducted a retrospective
Evans and coworkers53 suggested that there exists a survey to assess the demography, presentation, clinical
spectrum-like relationship between acquired course, and treatment response of children with Evans’s
hemolytic anemia and thrombocytopenic purpura; on syndrome. Information was analyzed from a detailed
the one hand, acquired hemolytic anemia with sensi- questionnaire completed by pediatric hematologists
tization of the RBCs is often accompanied with throm- mainly in the United States and Canada. The question-
bocytopenia, whereas, on the other hand, primary naire sought information regarding demographics,
thrombocytopenic purpura is frequently accompa- findings at presentation, approach to diagnosis, treat-
nied with RBC sensitization with or without ments used (with specific reference to splenectomy, cor-
hemolytic anemia. Indeed, in addition to these clinical ticosteroids, and intravenous immunoglobulin [IVIG]),
observations, Zucker-Franklin and Karpatkin54 pre- course of the disease with emphasis on recurrences, and
sented data indicating that autoimmune mechanisms status at last follow-up. Forty-two patients (22 males
may be directed against RBCs as well as platelets in and 20 females) were included in the study. The median
most patients with severe idiopathic autoimmune age was 7.7 years (range, 0.2 to 26.6 years). At presenta-
thrombocytopenia. They noted that routine blood tion, thrombocytopenia (32 patients) and anemia (28)
smears of patients with chronic autoimmune idio- were common; neutropenia occurred in 10 and pancy-
pathic thrombocytopenic purpura (ITP) commonly topenia in 6. Patients received a median of 5 (range, 0 to
showed platelets larger than normal with many giant 12) modalities of treatment. Courses of IVIG and corti-
forms, but Coulter-counter volume measurements of costeroids were given to almost all patients; responses
platelets also revealed a distinct peak in the area in were varied, but the effects lasted as long as 2 years.
which particles with volumes much below those of Splenectomy was performed for 15 patients, but the
normal platelets would be located. By electron median duration of response was only 1 month. Other
microscopy, RBCs as well as platelet fragments were treatments included cyclosporine, vincristine, danazol,
found in the 27,000 × g plasma sediment of 15 patients azathioprine, cyclophosphamide, and plasmapheresis.
with severe disease. These fragments were not The course of the disease was characterized by recur-
observed in the plasma sediment of 12 normal sub- rent thrombocytopenia, hemolytic anemia, and neu-
jects, 2 healthy asplenic subjects, 3 patients with tropenia. After a median follow-up of 3 years, 3 patients
thrombocytopenia of nonimmunologic origin, and 2 had died, 20 had active disease on treatment, 5 had
with ITP in remission. They concluded that, in addi- persistent disease (not on treatment), and 14 had no
tion to destroying platelets, their patients had subclin- evidence of disease. The authors concluded that Evans’s
ical hemolysis. syndrome is a chronic and recurrent condition that is
AIHA and thrombocytopenia do not necessarily often refractory to IVIG, corticosteroids, and splenec-
appear at the same time.2 A number of cases have tomy. Responses to other agents have been anecdotal
68 Immune Hemolytic Anemias

and inconclusive. They suggested that a prospective different response to immunosuppressive treatment.
study involving these agents is needed to determine The leukopenia of two and the thrombocytopenia of
optimal therapeutic combinations. six patients appeared at varied time intervals after the
Savasan and coworkers52 reported 11 patients (10 AIHA or the detection of leukocyte and platelet anti-
boys and 1 girl) with Evans’s syndrome with a median bodies. Thrombocytopenic purpura was present in
follow-up time of 8 years who were evaluated retro- six patients, in two of these since infancy. The authors
spectively. Six patients had either persistent hepato- suggested that AIHA may be a complex autoimmune
splenomegaly or generalized lymphadenopathy, or syndrome that may involve leukocytes and platelets
both. In five patients, an increase in lymph node as well as RBCs, with synthesis of autoantibodies
and/or spleen size was observed during the exacerba- specific for different blood cells.
tions of cytopenias. Seven patients had quantitative Pegels and coworkers60 used an immunofluores-
serum immunoglobulin abnormalities at the time of cence test for autoantibodies to study 24 patients with
presentation. There were associated systemic manifes- Evans’s syndrome and an additional 29 patients with
tations in nine patients. Various forms of treatment both ITP and idiopathic neutropenia (INP) but without
were used with mixed results. Four patients died from AIHA. The direct immunofluorescence test on platelets
sepsis and haemorrhage; four had complete recov- and/or on granulocytes was positive in all patients
ery⎯two after splenectomy. The authors concluded with a cytopenia, but the sera of only 17 patients with
that Evans’s syndrome is a heterogeneous disorder Evans’s syndrome and 15 of the other patients con-
with significant morbidity and mortality. There was a tained platelet- or granulocyte-specific autoantibodies.
high incidence of quantitative serum immunoglobulin From absorption and elution experiments, the authors
abnormalities, lymphoid hyperplasia, and associated concluded that the autoantibodies were directed
systemic manifestations, suggesting that Evans’s syn- against antigens specific for the various peripheral
drome may represent a stage of a more broad spectrum, blood cells, that is, RBCs, platelets, and granulocytes,
generalized immune dysregulation. and that they were not cross-reacting.
Crosby57 and Crosby and Rappaport35 have pointed Miller and Schultz Beardsley61 described three chil-
out that the prognosis appears worse when thrombo- dren with, concurrently or successively, neutropenia,
cytopenia is present in AIHA. Of the patients they thrombocytopenia, and AIHA. One of them devel-
reviewed, 12 of 17 patients with thrombocytopenia oped reticulocytopenia. The DAT was positive and
died, compared with 5 of 16 patients who did not have antiplatelet and antileukocyte antibodies were pre-
thrombocytopenia. Chertkow and Dacie58 derived a sent. In two of the children it was possible to show
similar conclusion from their data, but Allgood and that the anti-RBC and antiplatelet antibodies were
Chaplin9 found no relationship between initial separate entities that did not cross-react.
platelet count and mortality. The high death rates Chapman and coworkers62 described the develop-
referred to in the earlier reports of Crosby and co- ment of autoimmune thrombocytopenia followed by
workers and Chertkow and Dacie were derived from AIHA in a woman with measles. An IgM platelet
data regarding patients for most of whom the only autoantibody was detected using a fluorescent-
available treatment was blood transfusion or labeled antiglobulin technique. The thrombocytope-
splenectomy.7 Whether thrombocytopenia affects nia resolved spontaneously, although the platelet
prognosis using modern therapy is uncertain. autoantibody persisted and platelet survival
remained shortened, suggesting a compensated
IMMUNOPANCYTOPENIA thrombocytolytic state. An IgG granulocyte autoanti-
body was present transiently, although the patient
Some patients with AIHA have developed immune was never neutropenic. The AIHA was due to an IgM
leukopenia and thrombocytopenia as well. Although cold autoantibody (anti-I), which was active up to
the publication by Evans and Duane46 is generally 30°C, and an IgG warm autoantibody, which was
cited as a description of Evans’s syndrome, which is detectable only when she was severely anemic. After
defined as AIHA and thrombocytopenia, two of the an initial blood transfusion, the anemia resolved and
five such patients reported by these authors also had the RBC autoantibodies disappeared. The platelet,
leukopenia. Indeed, the authors suggested that the granulocyte, and red cell autoantibodies were cell
leukopenia and thrombocytopenia were probably due specific and not a single cross-reacting antibody.
to “an immune body” with a broader range of activity Wiesneth and coworkers63 investigated four patients
than the RBCs or to a separate immune substance or with combined immunocytopenia of unknown origin.
substances more specific for platelets and WBCs. Two patients with pancytopenia had alloantibodies and
Fagiolo59 studied platelet and leukocyte counts and autoantibodies against RBCs, granulocytes, and throm-
leukocytotoxic and platelet antibodies in 32 patients bocytes. Two other patients with granulocytopenia and
with AIHA. Leukopenia was present in 59.4%, throm- thrombocytopenia showed alloantibodies and autoanti-
bocytopenia in 59.4%, and leukothrombocytopenia in bodies against granulocytes and thrombocytes. All
40.5% of the cases. Specific antibodies for granulo- patients went into a transient or persistent remission
cytes were found in 81.3%, and platelet antibodies in under immunosuppressive therapy. The normalization
90.6%. The AIHA, leukopenia, and thrombocytopenia of peripheral blood counts correlated with the disap-
generally presented a dissociated evolution and a pearance of antibodies, suggesting that the cytopenias
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 69

TABLE 3-5. CLINICAL FEATURES OF FIVE PATIENTS WITH AIHA AND RETICULOCYTOPENIA

Hematocrit
on Duration of
Age (yr)/ Admission Reticulocytopenia Transfusion
Patient Sex (%) (days) (U) Outcome

1 52/F 10 10 19 Recovery
2 78/F 9 4 2 Recovery
3 53/F 10 90 53 Compensated status
4 39/M 9 8 5 Compensated status
5 49/F 8 160 84 Compensated status

From Conley CL, Lippman SM, Ness PM, Petz LD, Branch DR, Gallagher MT: Autoimmune hemolytic anemia with reticulocytopenia and
erythroid marrow. N Engl J Med 1982;306:281–286.

were caused by an antibody-mediated autoimmune finding in patients with WAIHA (see Chapter 2).
mechanism. However, reticulocytosis is not uniformly present
and, indeed, reticulocytopenia is recognized as an
important, although uncommon, manifestation of
BONE MARROW FINDINGS AIHA. Diagnosis is more difficult because AIHA does
not seem to be a logical consideration in the presence
As in other hemolytic anemias, the bone marrow nor-
of reticulocytopenia.
mally undergoes hypertrophy that is roughly propor-
Reticulocytopenia. Liesveld and coworkers44
tional to the intensity of the hemolysis.36 The
reviewed data on 109 consecutive patients with various
hypertrophy is primarily due to hyperplasia of ery-
types of autoimmune hemolysis, of whom 51 had
thropoietic cells, with the result that the erythroid/
WAIHA. The reticulocyte counts ranged from 0.4% to
myeloid ratio may even exceed unity. The erythro-
92% (Fig. 3-3). Twenty percent of the patients had an
poiesis is typically normoblastic in type and is usually
initial count of less than 4%, and 37% had an initial
present even in the presence of reticulocytopenia36,44
reticulocyte production index of less than 2 times basal.
(see later). With severe hemolysis, erythropoiesis
Eighty-eight cases had serial reticulocyte measure-
tends to become abnormal: In some cases there is a
ments, and in only 15% of patients did the reticulocyte
tendency for the nuclei of mature normoblasts to
production index remain less than 2 times basal. Thus,
break up into two or more lobes of varying size and
in most cases, the initially low reticulocyte production
for Howell-Jolly bodies to be present in the cytoplasm
index probably represented a lag in marrow respon-
of some of the normoblasts. Overt megaloblastic
siveness to hemolytic stress. However, in some cases,
erythropoiesis certainly occurs,9 as in other types of
the reticulocytopenia may be prolonged.36,64-71
hemolytic anemia, but it has not been reported often.
AIHA with reticulocytopenia should be considered
a medical emergency. Conley and coworkers64,72
RETICULOCYTES described five patients with AIHA in whom reticulo-
cytopenia was associated with life-threatening
As in other types of hemolytic anemias, a persistently anemia. Each of the patients had a hematocrit of 10%
raised reticulocyte count is a typical and characteristic or less on admission (Table 3-5). The patients survived

(70) (92)
100
(64) (79)

80
Cumulative percent

FIGURE 3-3. Cumulative percentage of first


uncorrected reticulocyte percentage count obtained at 60
diagnosis in 108 cases. Approximately 20% of patients
had an initial reticuloycyte count of less than 4%. (From
Liesveld JL, Rowe JM, Lichtman MA: Variability of the
40
erythropoietic response in autoimmune hemolytic
anemia: Analysis of 109 cases. Blood 1987;69:
820–826.)
20

0
0 4 8 12 16 20 24 28 32 36 40 44 100
Reticulocyte count (%)
70 Immune Hemolytic Anemias

because of prompt treatment with repeated transfu- crisis was associated with the presence of two distinct
sions, even though compatible cross-matches could antibodies, one directed at the mature RBCs and the
not be obtained. other at the erythroid progenitor cells. Dacie36 has
Numerous other case reports have emphasized proposed that reticulocytopenia with an erythroid
reticulocytopenia as an important factor in patients marrow may be caused by autoantibodies that react
with AIHA.36,65-68,73,74 The patient of Hauke and only with reticulocytes and mature RBCs.
coworkers66 required 38 units of RBCs over a period Parvovirus B19 and Reticulocytopenia. Parvovirus
of 24 days, during which the patient did not respond B19 infection has also been implicated as a cause of
to prednisone therapy. Splenectomy was performed; some cases of transient aplastic crises.86,87 Indeed, the
immediately afterward the peripheral RBC destruc- first clinically significant illness associated with B19
tion stopped, and further transfusions were not infection was hypoplastic crisis in patients with sickle
required. Greenberg and coworkers65 described two cell anemia.88 The erythroid tropism of B19 is due to
children with AIHA and reticulocytopenia that per- the tissue-specific expression of the cellular receptor
sisted for 165 and 40 days, respectively, despite for the virus. Binding is mediated through globoside,
aggressive treatment. Both children ultimately recov- which is found predominantly on erythroid cells or
ered fully. Olcay and coworkers67 and Carapella and their progenitors, where it is known as the blood
coworkers68 described similar cases. group P antigen.86,89,90 Only RBCs containing the
An early report by Crosby and Rappaport75 indi- P antigen can be hemagglutinated, and the bone
cated that 15 of 34 patients with idiopathic AIHA were marrow from patients that do not have P on their
reticulocytopenic and only 3 of the 15 patients sur- erythrocytes cannot be infected with parvovirus B19
vived. However, with modern therapy, such a poor in vitro. Susceptibility to parvovirus B19 increases
prognosis may not exist. Indeed, Allgood and with differentiation, and the pluripotent stem cell
Chaplin9 found no statistically significant relationship appears to be spared. The virus is directly cytotoxic,
between mortality and the initial reticulocyte count. with infected cells showing the ultrastructural and
Hyperplasia of the bone marrow is usually present in biochemical features typical of apoptosis.
patients with reticulocytopenia.36,44 Liesveld and Transient aplastic crises due to B19 infection have
coworkers44 found that 16 of 21 patients (76%) with a now been described in a wide range of patients with
reticulocyte production index of less than 2.0 had ery- underlying hemolytic disorders, including AIHA.80,82,91
throid hyperplasia. Only one patient had erythroid Parvovirus should be the presumptive diagnosis in
hypoplasia. All five patients reported by Conley and any patient with anemia due to abrupt cessation of
coworkers64 had an “intensely erythroid marrow” erythropoiesis as documented by reduced reticulo-
despite severe reticulocytopenia. Additional cases have cytes and the characteristic bone marrow morphology.
been reported by numerous authors.12,66,67,73,75-77 Patients with transient aplastic crises are often viremic
In other patients, AIHA and reticulocytopenia asso- at the time of presentation with high concentrations of
ciated with erythroid hypoplasia in the bone marrow the virus. As the B19 DNA is cleared from the serum,
have been reported.2,68,70,78-82 B19-specific IgM becomes detectable. The diagnosis
Mechanisms for Reticulocytopenia. A proposed therefore is readily made by detection of B19 DNA in
mechanism regarding the presence of reticulocyto- the serum or by the detection of specific antibodies of
penia and erythroid hypoplasia is the presence of an IgM class. Following the acute event, IgG confers life-
autoantibody that may or may not be distinct from long immunity to reinfection.86,87 Because aplastic
that acting on reticulocytes and mature RBCs. Expe- crises caused by parvovirus B19 typically last about
rimental studies suggest that antigenic determinants 7 to 14 days92 (except in patients with immune
on RBC precursors can react with erythrocyte auto- deficiency),89 it seems likely that prolonged episodes
antibodies. Pisciotta and Hinz83 and Yunis and Yunis84 of reticulocytopenia in AIHA are generally caused by
have demonstrated that RBC autoantibodies can inter- other mechanisms.36
act with nucleated RBCs. Further, Meyer and cowork-
ers78 described a patient with SLE and AIHA PROGNOSIS AND SURVIVAL
complicated by periodic episodes of red cell hypopla-
sia. These investigators demonstrated that erythroid The natural history of WAIHA is unpredictable, with
colony formation by normal human bone marrow no firm conclusions possible regarding clinical, hema-
cells in vitro was inhibited by both the patient’s serum tologic, or serologic features that would enable one to
and an eluate from his RBCs. Autoantibodies from predict the ultimate course in a patient once the
other patients with AIHA but without pure RBC diagnosis has been made. Some early suggestions
aplasia did not cause such inhibition. Mangan and that mortality is increased in patients who have a pos-
coworkers85 studied a patient with AIHA, reticulocy- itive indirect antiglobulin test10 or who have reticulo-
topenia, and RBC aplasia. They found that the cytopenia35 have not been confirmed in more recent
patient’s RBC autoantibody was a complement-inde- series.9,93,94
pendent IgG that reacted with the e antigen and that Early reviews on prognosis were discouraging. In
the patient also had a complement-dependent serum 1957, Crosby and Rappaport95 reported a 32%
IgG inhibitor directed against erythroid colony- and mortality rate in 50 patients, and in 1959 Dausset
burst-forming units. They suggested that the aplastic and Colombani10 reported a 27.7% mortality rate in
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 71

83 idiopathic cases. Pirofsky,2 in reviewing patients Cold Agglutinin Syndrome


studied between 1958 and 1966, reported a mortality
rate of 38% in patients with idiopathic AIHA and a INCIDENCE
mortality twice as great in secondary AIHA (primarily CAS is relatively uncommon compared with WAIHA,
associated with lymphoproliferative disorders). The but it occurs much more frequently than PCH. In our
mortality rate reported by Allgood and Chaplin9 was series of 347 patients with AIHAs (including drug-
38% in their 47 patients with idiopathic AIHA observed induced immune hemolytic anemia), 54 patients
between 1955 and 1965. Dacie94 reported that mortality (15.6%) had CAS (see Table 3-3). Dacie and Worlledge5
in 50 patients with idiopathic warm antibody AIHA reported that 25% of their patients with AIHA had CAS,
investigated between 1947 and 1961 was 46%. whereas van Loghem and coworkers100 and Dausset
Some more encouraging reports have appeared. In and Colombani10 reported an incidence of only 7.7%.
1974, Worlledge96 suggested that a better prognosis
may have resulted from improved therapy, because
the mortality was only 14% in 85 patients followed for AGE AND SEX DISTRIBUTION
periods of 3 months to 7 years. This finding compares
favorably with the earlier report cited previously94 Patients with idiopathic CAS are typically middle-
from the same institution in which the mortality rate aged or elderly. Worlledge101 reported that the
was 46%. Silverstein and coworkers93 analyzed the average age of 29 patients was 66. In Dacie’s series,
records of 117 patients observed between 1955 and only 3 of 21 patients with idiopathic CAS were
1965. Using actuarial survival curves, they concluded younger than 50, whereas 9 of 12 postpneumonia
that the lethality of acquired hemolytic anemia is cases occurred in patients between the ages of 20 and
much less than previously reported. The survival rate 45.102 Schubothe103 noted a peak incidence of onset
was 91% at 1 year, 76% at 5 years, and 73% at 10 years. from ages 51 to 60. However, 26% of the patients were
Poschmann and Fischer97 reported that the mortal- less than 41 years of age, a finding possibly resulting
ity rate of AIHA appears to be considerably lower in from the inclusion of patients with the acute transient
children than in adults. In their review of the litera- variant of the disease who are more frequently
ture, they found that the mortality in children was younger. Chronic idiopathic cold agglutinin disease
10% to 28%, whereas in adults, it was 28% to 70%. occurs only very rarely in childhood.13,102,104 In these
Nearly all children with an acute transient type of cases, there is usually evidence of a preceding viral
AIHA recover rapidly,13,98,99 and the difference in infection. Both sexes are affected, and there is no clear
prognosis may depend on the frequency of acute cases predominance in males or females.
and of associated malignant disorders. Acute self-
limited disease occurs in about 42% of children13 SYMPTOMS AND SIGNS
but in only about 4% of adults.97 Sokol and cowork-
ers,11 in their review of AIHA in childhood and ado- Symptoms are frequently just those of a chronic
lescence, reported that only 2 of 42 children had died; anemia that progresses gradually.105 In idiopathic
83% recovered, usually within 6 months. Habibi and cases, the disorder usually remains quite static and at
coworkers13 found that 80% of his pediatric patients worst only progresses slowly in intensity (Table 3-6).
had acute, transient disease with no deaths, but there The clinical manifestations vary greatly from patient
was an 11.2% mortality rate in chronic cases. For to patient, probably depending on the thermal range of
further details regarding prognosis in children, see the cold antibody. Clinical symptoms are more serious
Chapter 9. for patients whose cold agglutinin is active at higher
The prognosis in secondary cases of AIHA is temperatures.106 Temperatures of 30°C and lower are
often dependent on the outcome of the underlying normally attained in the superficial skin vessels of
disease.2 those parts of the body exposed to cold air or water.103
The thermal range of the antibody is more important
than the agglutination titer for clinical purposes.106,107
RECOVERY FROM AIHA Anemia is generally increased in wintertime as the
peripheral circulation is more likely to fall to a temper-
Extensive data are not available to establish a “cure” ature that facilitates the action of the antibody105 (see
rate for patients with AIHA. Dacie94 reported that Chapter 11, Fig. 11−9). Exposure to cold may precipitate
10 of 50 patients seen in the 1950s achieved clinical acute hemolytic episodes.106,108,109
and hematologic cure. However, in 3 of the 10, the Patients may experience hemoglobinuria, particu-
DAT was still positive when the patients were last larly in cold weather, and this finding is prominent
investigated, and whether any of the patients relapsed among manifestations listed in reviews of the disor-
later was unknown. Allgood and Chaplin,9 in their der. However, its incidence is difficult to determine,
review of 39 patients with warm antibody AIHA, all and in many patients hemoglobinuria is never ob-
over the age of 10 years, reported that 21 of them served. This is true even in some patients who de-
(54%) had regained normal hemoglobin levels and velop severe acrocyanosis during cold exposure.102,110
reticulocyte counts. In nine, despite clinical and hema- When an episode of intravascular hemolysis does
tologic recovery, the DAT remained positive. occur after cold exposure, it is not associated with
72 Immune Hemolytic Anemias

The acrocyanosis associated with cold agglutinins


TABLE 3-6. COLD AGGLUTININ SYNDROME
may be distinguished from the much more common
CHARACTERISTIC CLINICAL MANIFESTATIONS
Raynaud’s phenomenon, which may be idiopathic or
associated with diseases such as collagen vascular dis-
Symptoms and Signs
orders.105 In the former, the exposed distal fingers or
Symptoms of chronic anemia toes turn a dusky blue that either quickly reverts to
Fatigue normal on warming or progresses to blanching after
Dyspnea on exertion more prolonged or more severe cold exposure. In
Weakness Raynaud’s phenomenon, the digits characteristically
Dark urine (especially in cold weather)
Acrocyanosis (occasionally leading to gangrene of digits) turn white from vasospasm and then, as ischemia is
Pallor and jaundice resolving, turn blue; this change may be followed by a
Hepatosplenomegaly and lymphadenopathy may be present hyperemic phase in which the exposed parts become
reddened. Further, exposure to moderate degrees of
Laboratory Findings
cold in patients with cold agglutinins will cause all
Mild to moderate anemia digits to be equally affected, whereas Raynaud’s
Prominent autoagglutination, especially at cold temperatures phenomenon not associated with cold agglutinins
Abnormal red blood cell morphology may cause marked blanching of one or two fingers
Modest degrees of spherocytosis, anisocytosis, poikilocytosis, with the others remaining normal.
polychromatophilia
Reticulocytosis Cutaneous necrosis is an unusual manifestation of
Jaundice CAS.121-124 The patient reported by Stone and cowork-
Hemoglobinuria ers121 developed skin necrosis at three different sites
Erythroid hyperplasia in the bone marrow of intravenous infusion that had been used for RBC
and saline infusions. Cryoglobulin and cryofibrinogen
test results were negative. Other unusual manifesta-
fever, chills, or acute renal insufficiency.2,103 This phe- tions include peripheral neuropathy that resolved
nomenon contrasts with the marked constitutional after successful treatment of the hemolysis with corti-
symptoms associated with hemoglobinuria in PCH costeroids and plasma exchange125 and multiple pul-
or in those few patients with WAIHA who have monary embolisms, which was attributed to the
hemoglobinuria. intravascular hemagglutination in a patient who
Many affected patients complain of acrocyanosis developed a cold agglutinin titer of 2048 and hemoly-
of the ears, the nose tip, fingers, and toes in cold sis during a Mycoplsma pneumoniae infection.31
temperatures, a condition that vanishes quickly on Other physical findings are not prominent. Pallor and
warming.106,111 These symptoms are caused by auto- jaundice may be present, and the intensity of these
agglutination of the patient’s RBCs in the skin capil- findings depends on the rate of hemolysis and the
laries, causing local blood stasis. As blood flows ability of the liver to excrete bilirubin. Hepatosple-
through the capillaries of the skin and subcutaneous nomegaly and lymphadenopathy are not prominent
tissue, the temperature of the blood falls to as low as findings.106 Dacie102 indicates that the spleen is palpably
28°C if the ambient temperature is low.112 In vivo enlarged only in a minority of patients, whereas
agglutination can be directly demonstrated by micro- Schubothe103 states that usually the spleen and some-
scopy of conjunctival vessels after local administration times the liver are slightly enlarged. Dausset and
of ice-cold saline or by finger plethysmography after Colombani10 noted splenomegaly in 6 of 10 patients.
local exposure to cold.113 Thomas and coworkers125 reported a patient with CAS
Occasionally, actual gangrene, particularly of a digit who had a sensory-autonomic polyneuropathy that re-
or digits, has been observed.114,115 Although the solved after the hemolytic anemia went into remission.
occurrence of gangrene after prolonged exposure to
cold would appear to be a logical consequence of vas- LABORATORY FINDINGS
cular insufficiency caused by autohemagglutination,
there is some question as to whether the presence of Autoagglutination of anticoagulated blood samples
cryoglobulins may not be an additive factor. In a that occurs quickly as blood cools to room temperature
minority of patients with CAS, cryoglobulinemia may is characteristic and frequently the first observation
be an associated finding and/or the cold antibody made to suggest the diagnosis. Autoagglutination is
may be a cold precipitable protein.107,115-118 Ferriman intensified by cooling the blood to 4°C and is reversed
and coworkers,110 Rorvik,119 and Gaddy and by warming to 37°C. This property distinguishes cold
Powell120 all reported cases of CAS associated with autoagglutination from rouleaux formation and also
tissue gangrene in which cryoglobulins were present. from autoagglutination, which occurs in some patients
However, Mitchell and coworkers114 specifically with WAIHA. In the latter case, the autoagglutination
pointed out that cryoglobulins were not present in the does not resolve at 37°C. The agglutination creates
patient they reported who developed gangrene affect- problems in making satisfactory blood films and per-
ing the distal phalanx of the left third toe and ischemic forming blood counts.106,126-129 However, it should be
changes of two right toes. stressed that clinically insignificant cold autoantibodies
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 73

will also cause autoagglutination. Such antibodies have finding contrasts strikingly with hemoglobin values
a low thermal amplitude and do not cause hemoly- in WAIHA or PCH.
sis.130 Therefore, the mere observance of cold auto- Schubothe103 describes the course of the disease as
agglutination is not diagnostic of cold agglutinin “monotonous,” and patients may survive many years,
disease, and the cold antibodies must be further char- with minimal disability. Most patients tolerate their
acterized, as described in Chapters 5 and 6. A rather mild or moderate anemia quite well. In more severe
common diagnostic error is overdiagnosis of CAS in cases, however, death may ensue from complications
patients who have benign cold antibodies and patho- of slowly progressive anemia or from complications
genic warm autoantibodies. of blood transfusion. Sometimes, after a considerable
RBC morphology is less abnormal than in patients time, a generalized malignant lymphoproliferation
with WAIHA, with lesser degrees of anisocytosis and may occur that is manifested in the bone marrow mor-
poikilocytosis.102 Similarly, spherocytosis is less intense phology, as described earlier. Occasionally, an inten-
but has been noted by Leddy and Swisher,131 Schreiber sification of hemolysis with an increase of the thermal
and coworkers,132 and Rosse.133 Leukocyte and platelet amplitude up to the range of the internal body tem-
counts are usually normal. Bilirubinemia of mild degree perature seems to lead to a fatal complication.
is common. Reticulocytosis usually is in proportion to Evans and coworkers140 described a patient with
the severity of hemolysis. However, reticulocytopenia typical CAS with acrocyanosis, hemoglobinuria, and a
has been reported,85,134 and, in some patients, this cold agglutinin titer of 16,000. He had an IgM mono-
occurs in the presence of a hyperplastic erythroid bone clonal protein evident on serum protein electrophoresis
marrow indicating ineffective erythropoiesis.135,136 and had a series of remissions and exacerbations of his
Also, a case of B19 parvovirus−induced red cell aplasia hemolysis. Nevertheless, his course was rather benign
complicating acute CAS has been reported,134 similar to over a 20-year period until he was lost to further follow
cases of WAIHA as described earlier. up. The authors commented that they thought his
Bone marrow examination shows a variable degree abnormal immunoglobulin production was benign.
of erythroid hyperplasia and a slight but definite In contrast, there are several reports of fatal cases of
increase of lymphoid cells.106 Several authors have CAS.141-143 In each case, the cold antibody was present
reported extensive lymphoid proliferation in some in low-titer at 4°C but had a high thermal amplitude.
cases, especially in the terminal stages of the In two of the patients the antibody reacted up to at
disease.103,137,138 Schubothe103 performed serial exam- 37°C,141,142 and, in the other case, the antibody was
inations of the bone marrow with quantitative differ- strongly reactive at 30°C, although nonreactive at
ential counts in 14 patients. The number of lymphoid 37°C.143
cells was between 10% and 24% in 5 cases, between
26% and 33% in 3 cases, and between 41% and 95%
MANAGEMENT
in 6 cases. In several patients, the lymphoid cells
increased during the course of the disease. He Therapy, as reviewed in detail in Chapter 11, often has
concluded that the disease belongs to the category of little effect on the outcome of the disease. Ulvestad
lymphoproliferative disorders and that its bone and coworkers139 reported a series of 15 patients, of
marrow cytology was similar to that of the macroglob- whom 6 had received chlorambucil or cyclophos-
ulinemia of Waldenström. phamide. Concentrations of IgM decreased after treat-
Ulvestad and coworkers139 found that serum C3 ment but there was no clinical improvement in the
was decreased in 9 of 15 patients and serum C4 was patients’ disease. Transfusion therapy is reviewed in
decreased in 11 patients. In seven patients C4 was Chapter 10, and the management of patients who
below the detection limit (<0.06 g/L). Six of these have cold agglutinins and who require surgery under
patients had a reduced complement level (CH50). hypothermia is reviewed in Chapter 9.
There was no statistical association between comple-
ment concentrations and anti-I/i titers.
Paroxysmal Cold Hemoglobinuria
COURSE AND PROGNOSIS INCIDENCE
The prognosis in patients with idiopathic CAS is Many reports emphasize that PCH is an unusual
significantly better than it is in patients with WAIHA. disease. Howard and coworkers144 noted only two
Most commonly, the patient has a chronic, fairly stable occurrences in 38 years and 298,878 admissions at the
anemia that is mild or moderate in severity. For Montreal Hospital. Becker145 noted that the diagnosis
example, in only 2 of the 12 patients reviewed by was made in only one patient in 20 years at the
Ferriman and coworkers110 did the hemoglobin fall University of Chicago clinic, during which time 130,000
below 7.4 g/dL. Similarly, Schubothe103 reported 16 patients were admitted to the hospital and 382,799
patients followed for “several years,” and every patients were seen as outpatients. Pirofsky2 encoun-
patient had a hemoglobin of 9 g/dL or greater at some tered only two occurrences over a I5-year period at
time during the period; in only 2 patients did the Bellevue Hospital in New York and at the University of
hemoglobin fall below 6.5 g/dL at any time. This Oregon hospitals and clinics. PCH was diagnosed in
74 Immune Hemolytic Anemias

only 4 of 34 children with acute AIHA by Habibi13 and antibiotics, syphilis was considered an etiologic factor
only 1 of 17 children reviewed by Buchanan and in a large majority of cases. Donath and Landsteiner’s
coworkers.12 Subsequent reports suggested a some- review of the literature from 1906 to 1925 revealed
what higher incidence. Dacie and Worlledge5 reported evidence of syphilis in 95 of 99 reported cases.156
15 cases out of a total of 195 patients with AIHA (5.1%), Becker145 and Nabarro157 also stressed this relation-
and van Loghem and his associates100 reported an inci- ship and pointed out that, when related to syphilis,
dence of 10% in their 168 patients. Johnsen and cowork- PCH appeared only in congenital syphilis or in the
ers146 encountered three cases within a period of quiescent stage of late syphilis. In these patients, PCH
7 months; especially remarkable is the report of Bird was a chronic comparatively benign disease with
and coworkers,147 who diagnosed this disorder in three exacerbations related to cold exposure.
children within a period of just 16 days. However, as early as 1908 Donath and Landsteiner
In more recent years, PCH has become recognized suggested that the disease might be caused by infec-
as being a relatively frequent cause of acute transient tions other than syphilis,158 and recent reports gener-
AIHA in childhood. Sokol and coworkers11 stated that ally describe an acute transient hemolytic anemia often
17 of 42 (40%) of patients less than 16 years of age with associated with a upper respiratory infection or a
AIHA had formed Donath-Landsteiner antibodies, “flu-like illness.” Most commonly, a specific infectious
and Göttsche and coworkers148 reported an incidence disease is not present but rather an infection of
in children with AIHA of 32% (22 of 68). Wynn and undefined etiology. Also, PCH has rarely been associ-
coworkers149 reported six cases of children presenting ated with other disorders, including non-Hodgkin’s
with PCH occurring in a 3-year period in the north- lymphoma159,160 and oat cell carcinoma.161 Some
west of England, and Lambert and coworkers150 patients have no history of an antecedent illness,
reported six episodes of “biphasic hemolysis” in four and they appear healthy until the onset of the acute
children over a 1-year period. Indeed, both paroxysm.147
Nordhagen and coworkers151 and Heddle152 have Infections and Paroxysmal Cold Hemoglobinuria.
suggested that PCH is one of the most common causes PCH has been associated with a very wide range of
of acute AIHA in young children. The reason why infectious agents. In the nineteenth and early twentieth
acute transient PCH appears to be a more common centuries, PCH was frequently found in association
type of childhood AIHA than it was several decades with latent syphilis, but with the development of effec-
ago is uncertain, but it probably relates to greater tive antibiotic therapy for syphilis, this association has
awareness of the disorder and more frequent use of become rare,153,155 although not unknown.155,162-168
the Donath-Landsteiner test, especially in children Kumar and coworkers168 studied 26 patients with
with acute AIHA with hemoglobinuria. syphilis (4 primary and 22 secondary syphilis) to
determine the incidence of PCH in this population.
CLASSIFICATION Three patients gave a history of passing dark urine,
although none of these presented with evidence of
Dacie153 has suggested that PCH should be classified acute hemolysis. The Donath-Landsteiner test was
into three categories: chronic syphilitic, chronic non- positive in 7 of the 22 patients with secondary syphilis
syphilitic, and acute transient. In modern times, (32%), including the 3 with a history of dark urine.
chronic PCH is very rare and almost all cases are of Anti-P specificity was also demonstrated in these
the acute and transient type. PCH may also be cases. In four patients, the DAT was positive with
classified as idiopathic or secondary. polyspecific and anti-C3 antisera but negative with
anti-IgG. Reticulocyte counts ranged from 2.5% to 7%
RACE, SEX, AND AGE DISTRIBUTION (mean, 4.92%) in the Donath-Landsteiner−positive
There does not appear to be any particular racial dis- cases. All patients were treated with penicillin over a
tribution for the development of PCH. Puzzling infor- period of 21 to 28 days. In three of the seven patients
mation is the fact that combined data in three with a positive Donath-Landsteiner test, the antibody
reviews148,152,154 indicate that 52 of 77 children were disappeared immediately after treatment, and in the
boys, for a boy/girl ratio of 2.1:1. others it decreased in titer or remained unchanged.
Acute PCH typically presents in young children. In The authors suggested that detection of the Donath-
the series of Sokol and coworkers,155 the median age Landsteiner antibody may have therapeutic implica-
at presentation was 5 years. In one study, all of 22 tions in syphilitic patients, especially in avoiding
patients with PCH were less than 9 years old; no cases exposure to cold temperatures, which can lead to
being seen among 531 adults with AIHA.148 Although paroxysms of hemolysis.
particularly common in children, the disease affects PCH frequently has its onset after a “viral syn-
patients of all ages, and the patients in the series of drome” of undetermined etiology153,155 and, in addi-
Sokol and coworkers155 ranged from 1 to 82 years. tion, has been found in association with the following
well-defined infections: measles,169,170 mumps,171
IDIOPATHIC AND SECONDARY TYPES chickenpox,11,172 coxsackievirus A9,173 parvovirus,174
cytomegalovirus (CMV),175 infectious mononucleosis
The relationship of PCH to syphilis was described (Epstein-Barr virus [EBV]),176-178 Haemophilus
more than a century ago and, until the discovery of influenzae,175 Mycoplasma pneumoniae,179,180 Klebsiella
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 75

pneumonia,181 adenovirus,154 influenza virus A,154 and TABLE 3-8. SUMMARY OF THE FREQUENCY
measles vaccine.169 OF SIGNS AND SYMPTOMS IN 42 REPORTED
There was considerable interest in the relationship CASES OF ACUTE TRANSIENT PAROXYSMAL
between PCH and parvovirus B19 infection after the COLD HEMOGLOBINURIA
discovery that the cellular receptor for the virus was
the erythrocyte P antigen182 and that people without No. of
the P antigen (pp and Pk phenotype) were resistant Sign/Symptom Reports
to the infection.89 It was suggested that parvovirus
could be implicated in the pathogenesis of PCH.182 Hemoglobinuria 41
However, Sharpe and coworkers183 tested the sera Jaundice 33
Pallor 28
from 13 patients with PCH and from 18 controls. Fever 23
Seven of the patients with PCH and 11 of the controls Abdominal pain 17
had anti-B19 antibodies of the IgG class, but none had Malaise 14
specific IgM antibodies indicative of recent infection. Cough 13
In contrast, Chambers174 did find both IgM and IgG Palpable liver/spleen 12
Anorexia 9
antibody against parvovirus and suggested that the Vomiting 9
infection could have been responsible for the reticu- Rigors 7
locytopenia and erythroid hypoplasia which were Chills 4
present in her patient. Headache 2
Loin pain 2
Diarrhea 1
SYMPTOMS AND SIGNS Back pain 1

The typical presentation is that of a child who, during From Heddle NM: Acute paroxysmal cold hemoglobinuria. Transfus Med Rev
1989;3:219–229.
the preceding 1 to 2 weeks, had experienced an upper
respiratory tract infection152,153,155,175 (Table 3-7).
Usually, the onset of hemolysis is signaled by a recur-
rence of fever and then the passage of red-brown were also common findings. Approximately 25% of
urine.153 Hemoglobinuria, jaundice, and pallor were cases presented with palpable liver and spleen.
the most common clinical findings in 42 cases of acute Hemoglobinuria after exposure to cold is rare in acute
PCH reported by Heddle152 (Table 3-8). Of particular PCH and was seen in only 1 of 52 patients reported by
note is that hemoglobinuria was found in 41 of the Sokol and coworkers155 Nevertheless, hemoglobin-
42 cases in that report. Abdominal pain and fever uria may be induced by exposure to cold.153,155

HEMATOLOGIC FINDINGS
TABLE 3-7. PAROXYSMAL COLD
HEMOGLOBINURIA—CHARACTERISTIC CLINICAL Hemoglobin. Severe and rapidly progressive anemia
MANIFESTATIONS frequently occurs. Lambert and coworkers150 reported
that their patient’s hemoglobin value decreased from 10
Symptoms and Signs to 3.7 g/dL over a 6-hour period, and Lindgren and
coworkers184 reported a fall in hemoglobin level in their
Patient usually a child patient from 14.1 to 3.2 g/dL in 3 days. In the three
History of a recent upper respiratory or “flulike” illness
Acute onset of illness
patients reported by Johnsen and coworkers,185 the
Fever hemoglobin of the least severely affected child was 9.8
Malaise g/dL at 1 day after the onset of symptoms, with a drop
Abdominal pain to 6.8 g/dL on the fourth day of the disease. A second
Red or red-brown urine is frequently present child had a hemoglobin level of 7.3 g/dL 1 day after the
Jaundice
Pallor onset of hemoglobinuria; despite being kept warm, the
hemoglobin fell to 4.7 g/dL 2 days later. The third
Laboratory Findings patient reported by these authors had a hemoglobin of
12.5 g/dL 2 days after the onset of shaking chills,
Anemia
Often severe
abdominal pain, and vomiting and the passage of red-
May be rapidly progressive brown urine. Hemoglobinuria persisted, and 3 days
Reticulocytosis (reticulocytopenia in some patients) later the hemoglobin was 5 g/dL, with a further drop to
Abnormal red blood cell morphology 4.2 g/dL 4 days later.
Spherocytosis, anisocytosis, poikilocytosis, autoagglutination, Levels of hemoglobin as low as 5 g/dL or
polychromatophilia
Erythrophagocytosis by neutrophils is commonly present even lower have been quite commonly re-
Hemoglobinuria (may occur on exposure to cold in the rare patient ported.169,170,175,183,186 Sokol and coworkers155
with chronic paroxysmal cold hemoglobinuria) reported that the minimum hemoglobin level had
Erythroid hyperplasia or normal results in bone marrow been less than 5 g/dL in 17 of 51 cases (33%), and
Leukocytosis (leukopenia in some patients)
Platelet count usually normal or elevated
Heddle187 reviewed 42 published cases and found
that the admission hemoglobin ranged from 2.5 to
76 Immune Hemolytic Anemias

12.5 g/dL with a mean value of 6.8 g/dL. Göttsche When it is observed, monocytes, not neutrophils, are
and coworkers148 reported hemoglobin levels of less more often involved,195 although erythrophagocytosis
than 5 g/dL in 6 of 22 cases, with the minimum being by neutrophils has been described in a patient with
4.4 g/dL. hemolytic anemia seemingly induced by cefotetan.196
Reticulocytes. A relative reticulocytopenia occurs It is understandable why IgG-mediated interactions
in some patients with PCH at the time of presenta- are not usually observed, as free IgG in the plasma is
tion.150,155,170,175,188 Heddle152 reported that reticulo- known to inhibit reactions between RBC-bound IgG
cytopenia was prominent feature in four of the five and the Fc receptor of monocytes. Normal plasma does
cases seen in her medical center. not contain the activation products C3b or iC3b, so
Red Blood Cell Morphology. The presence of sphe- monocyte/neutrophil interactions with RBC-bound
rocytes, anisocytosis, poikilocytosis, fragmented red C3b or iC3b are not inhibited. Thus, RBCs coated with
cells, basophilic stippling, polychromatophilia, auto- C3b can react with the CR1 receptor of monocytes and
agglutination, erythrophagocytosis, and nucleated neutrophils, and iC3b (the first breakdown product of
erythrocytes have all been reported.2,155,175,185,189 C3b) can react with the CR1 and CR3 receptors. The
Spherocytosis is particularly common in the peripheral final C3 breakdown product, C3d (C3dg), cannot react
blood of severely affected patients.147,153,155,170,175,185 with monocytes or neutrophils as there are no C3d
Osmotic fragility is increased commensurate with the receptors on monocytes or neutrophils.
spherocytosis present153 but has seldom been Thus, although it is unclear how neutrophil ery-
recorded. throphagocytosis takes place in PCH, or its clinical
Bone Marrow Examination. Bone marrow aspira- significance, the presence of this phenomenon, as
tion is not often performed, but, when it is carried out, emphasized in several reports,152,181,190,197 should alert
it generally reveals normal results or erythroid hematopathologists to the possible presence of PCH.
hyperplasia.155 Leukocytes. Leukopenia followed by a neutrophil
Erythrophagocytosis. Red cell adherence and ery- leukocytosis with a shift to the left may occur during
throphagocytosis by neutrophils, rather than mono- a paroxysm induced by cold. Leukopenia was noted
cytes, is a prominent finding in acute transient as early as 1913, and remarkable drops in the WBC
PCH153,190 but is seen rarely in other forms of count have been recorded, particularly in the older
immune-mediated hemolytic anemia,152 including medical literature, regarding patients with chronic
WAIHA.190 Dacie153 mentioned that this phenomena PCH who have been exposed to cold.153 Uchida198
was first noted by Ehrlich in 1891 and that it had been found that maximum leukopenia occurred 5 to 20
reported repeatedly in subsequent years. Heddle152 minutes after chilling, with the fall being as much as
reported the phenomenon to be present in 20% of all 72%. After about 2 hours, leukocytosis developed.
PCH cases and in 80% of young children with acute Similaily, Montagnani199 described a reduction of
transient PCH. She suggested that the phenomenon leukocytes from 9800 to 1000/μL in an artificially pro-
may be underreported as it is transient; she described duced attack.
a case where erythrophagocytosis by neutrophils was The modern literature usually describes normal
noted on the day of admission but was not seen on the WBC counts or leukocytosis during acute attacks. In
next day.152 There are three reports devoted to this the 52 patients with acute PCH investigated by Sokol
phenomenon; each of these reports is accompanied by and coworkers,155 leukocytosis was present in 24
photographs of erythrophagocytosis191-193 (Fig. 3-4). patients and varied from mild to severe. Göttsche and
It is unclear why the presence of neutrophil- coworkers148 reported counts of between 10,500 and
mediated erythrophagocytosis on the peripheral blood 38,600/μL in 21 patients, with nine of the counts
smear is associated so commonly with PCH. It is also exceeding 30,000/μL.
unclear whether neutrophil-mediated RBC destruction Platelets. Platelet counts seem to have been
may add to the hemolytic process in vivo. Garratty and carried out infrequently in cases of acute PCH.153
coworkers194 noted that DAT-positive RBCs from PCH Where reported, the counts usually were normal,
patients are phagocytosed by monocytes in an in vitro although increased numbers have been reported in a
monocyte monolayer assay, in contrast to DAT-positive few patients. Sokol and coworkers155 found throm-
RBCs from patients with cold agglutinin disease. This bocytosis in 12 of his 52 patients. Göttsche and
is a surprising finding as in both conditions RBC- coworkers148 performed counts in 20 patients at
bound C3d, but no immunoglobulin (i.e., “cold” IgM or presentation and found slight thrombocytopenia
IgG antibodies), is detected by the DAT, and there are (139,000/μL) in 1 patient, with the counts in the
no C3d receptors on monocytes/macrophages. It was other patients ranging from 210,000 to 559,000/μL.
suggested that perhaps RBCs from PCH are sensitized
with small amounts (below threshold of the DAT) of LABORATORY DIAGNOSIS
the IgG Donath-Landsteiner antibody and that C3d
may be synergistic for the monocyte RBC-bound IgG Serologic tests necessary for the diagnosis of PCH are
interaction.194 described in Chapters 5 and 6, and specificities of the
Erythrophagocytosis is rarely observed in the antibodies found in this disorder are reviewed in
peripheral blood smear of the more common WAIHA. Chapter 7.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 77

FIGURE 3-4. Paroxysmal cold hemoglobinuria (PCH) is a hemolytic anemia characterized by a biphasic
hemolysin. Most cases involve children and are typically fulminant, postinfectious, and self-limited. Adult
cases are uncommon and typically involve a chronic or relapsing course. In this case, a 59-year-old white
woman experienced two episodes of anemia over 2 months in the winter of 1999. Transfusion attempts
were stymied because of in vitro hemolysis observed in LISS antibody screens. PEG-IAT antibody screens
were nonreactive. Her peripheral blood smear revealed striking red blood cell (RBC) rosetting around
neutrophils (1) and rare erythrophagocytosis in neutrophils and monocytes (2). This prompted the
performance of an indirect Donath-Landsteiner test, which demonstrated a biphasic hemolysin leading to
the diagnosis of PCH (3). In 3, hemolysis is seen in Tubes A1 (patient’s serum + donor RBCs) and A2
(patient’s serum + fresh normal serum + donor RBCs), but not in Tube A3 (fresh donor sera + donor
RBCs) after being incubated initially at 4°C and then warmed to 37°C. Similar tubes held at 4°C or 37°C
did not show hemolysis. Donath-Landsteiner testing should be considered whenever RBC adherence to
neutrophils or erythrophagocytosis is seen in peripheral blood smears from anemic patients. (From
Depcik-Smith ND, Escobar MA, Ma AD, Brecher ME: RBC rosetting and erythrophagocytosis in adult
paroxysmal cold hemoglobinuria. Transfusion 2001;41:163.)

CHRONIC PAROXYSMAL COLD


HEMOGLOBINURIA the onset of symptoms, and the interval between chill-
ing and the development of symptoms ranged from a
Both the syphilitic and nonsyphilitic forms of chronic few minutes to 8 hours. The extent of cold exposure
PCH are presently reported only exceedingly rarely. could be surprisingly slight, and, in some cases, a
The older medical literature describes chronic history of undue exposure to cold was not elicited. The
syphilitic PCH as a rather benign disease that rarely requirement for cold exposure probably depended on
caused severe chronic anemia. However, acute attacks the thermal amplitude of the Donath-Landsteiner anti-
of hemolysis and hemoglobinuria were well known body. In patients in whom cold exposure was docu-
and were characterized by the sudden onset of shaking mented, the patient’s temperature rose to as high as
chills, fever, malaise, abdominal distress, aching pains 40°C (104°F). The acute constitutional symptoms and
in the back or legs, and nausea. Usually, hemoglobin the hemoglobinuria usually resolved within a few
was present in the first specimen of urine passed after hours but could persist for days.
78 Immune Hemolytic Anemias

Not all of the preceding findings were present in specificity of the antibody was shown to be typical of
each patient or in each paroxysm occurring in a given that of Donath-Landsteiner antibodies, namely anti-P.
patient. Hemoglobinuria occurred in some patients The antibody caused agglutination as well as lysis; at
without other acute manifestations, and, conversely, 20°C the indirect antiglobulin test (IAT) was 1+ with an
constitutional symptoms in the absence of overt anti-IgG serum and 4+ with an anti-C serum.
hemoglobinuria have been described. Nelson and
Nichol165 pointed out that constitutional symptoms TREATMENT AND PROGNOSIS
associated with attacks of hemoglobinuria in PCH are
generally more severe than when patients who have Transfusion therapy of PCH is reviewed in Chapter 10,
CAS develop hemoglobinuria in cold weather. This and medical management is reviewed in Chapter 11.
phenomenon is probably related to the sudden inten- The prognosis of acute PCH is excellent, although
sive of hemolysis in PCH, which may result from the deaths have been reported.154 PCH is rarely a cause
remarkable hemolytic potency of the Donath- of severe chronic anemia or death. However, acute
Landsteiner antibody. attacks are frequently severe, as has been indicated.
There are many descriptions in the older medical The acute illness characteristically resolves sponta-
literature of Raynaud’s phenomenon (more accu- neously within a few days to several weeks after onset
rately, acrocyanosis) occurring in association with and does not recur,175 although Lambert and cowork-
attacks of hemoglobinuria in probable cases of ers150 described relapses in two patients 9 to 10
PCH.153 The fingers, toes, tip of the nose, lips, or ears months after the initial hemolytic episode⎯one after
may be blanched or become deeply cyanotic, and even a cold exposure (+14°C) and the other without evi-
gangrene has been described. dence of any causal agent.
Although chronic nonsyphilitic PCH is exceedingly The Donath-Landsteiner antibody may persist in low
rare, a number of case reports have appeared in the titer and with low thermal amplitude for a longer
medical literature.153,181,200-206 A particularly interest- period of time. Indeed, Dacie reported the persistence
ing report is that of Ries and coworkers,200 who of the Donath-Landsteiner antibody for 8 years after a
described a 60-year-old patient who had a chronic single episode of hemoglobinuria.207 More commonly,
moderately severe hemolytic anemia. Hemoglobinuria the titer and thermal range of the antibody fall rapidly,
occurred intermittently but did not seem to be related so that 2 to 3 months later it is no longer detectable.96
to cold exposure. The Donath-Landsteiner test was Chronic PCH secondary to syphilis generally has a
positive, and tests of the in vitro thermal amplitude of long, comparatively benign course,153,200 although
the antibody indicated that sensitization could take there are reports of two patients with chronic PCH
place at temperatures ranging from 0°C to the excep- who developed peripheral gangrene.155 There are
tionally high temperature of 32°C. Lysis also took place sporadic reports of fatalities, but these generally
readily in monophasic tests at temperatures from 10 to occurred in elderly patients with a variety of under-
32°C; the optimum was 15°C to 20°C (Fig. 3-5). The lying conditions.155

1:128 Indirect antiglobulin


test (anti-C3)
Biphasic hemolysis
1:64
Hemagglutination
Monophasic hemolysis
1:32

1:16
Serum dilution

FIGURE 3-5. Temperature requirements for lysis,


agglutination, and the IAT given by a D-L antibody of
1:8
exceptionally high thermal activity. (From Ries CA, Garratty
G, Petz LD, Fudenberg HH: Paroxysmal cold hemoglobinuria:
Report of a case with an exceptionally high thermal range
1:4
Donath-Landsteiner antibody. Blood 1971;38:491–499.)

1:2

1:1

0⬚ 5⬚ 10⬚ 15⬚ 20⬚ 25⬚ 30⬚ 32⬚ 35⬚ 37⬚


Temperature (centigrade)
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 79

Combined Cold and Warm AIHA readily demonstrable by IAT using serum and/or
or “Mixed” AIHA eluate, and, in tests against a routine panel of RBCs, no
specificity was evident. Three of the four patients were
In unusual instances, a patient is found who has diagnosed as having an associated lymphoproliferative
serologic findings characteristic of WAIHA while also disorder, but in each instance this diagnosis was based
having a cold agglutinin of high titer and thermal only on the finding of excessive numbers of lympho-
amplitude, thus satisfying the criteria for both warm cytic cells and plasmacytoid lymphocytes in the bone
antibody AIHA and CAS.1 Such cases are frequently marrow. None of the patients had other manifestations
referred to as “combined cold and warm AIHA,” or of lymphoma such as lymphadenopathy, and, in parti-
“mixed” AIHA, and reviews of AIHA generally cular, laparotomy and splenectomy in two patients
include such a category.208-213 failed to reveal evidence of lymphoma. All patients
responded to therapy during the period of follow-up
CASE REPORTS that ranged from 2 weeks to 2 years.
Sokol and coworkers216 described 25 patients with
Crookston was among the first to provide a detailed mixed AIHA and indicated that such patients
report of such a case.214 His patient was a 70-year-old accounted for about 7% of their cases of AIHA. In all
man with a lymphoproliferative disorder who devel- patients, the DAT was positive and the RBCs were
oped AIHA and presented with a hemoglobin level of coated with IgG and complement. No cold agglutinin
4.7 g/dL. The reticulocytes were 5%; WBC count, titers were reported, but all patients had cold autoan-
6500/μL with a normal differential count; and platelets, tibodies that were active at 30°C or above. The cold
130,000/μL. The blood film showed moderate autoag- autoantibodies often showed blood group specificity
glutination and marked spherocytosis. A bone marrow within the Ii system; specificity was not usually
biopsy showed a very hypercellular specimen almost evident in the warm component. Hemolysins were
completely replaced by small lymphocytic cells. A found in 18 patients. Serologic reactions generally
sharp “M” spike was present on serum electrophoresis remained mixed throughout the course of the illness,
and the paraprotein was identified as an IgM protein although cold antibody was eventually found alone in
containing only λ light chains. one patient and, in four others, warm antibody alone
The DAT was strongly positive with anti-IgG and emerged. Moderate to severe thrombocytopenia was
anti-C3 antiglobulin reagents. The patient’s serum present in four patients, and in three others an occa-
contained anti-E and anti-K alloantibodies, but a sional low platelet count was found. Mixed AIHA was
warm IgG autoantibody was also identified in his found in all age groups but was more frequent in later
serum and eluate. The autoantibody reacted at 37°C life; the male/female ratio was 1:1.5. The cases were
with all red cells tested except Rhnull cells. The patient either idiopathic (44%) or secondary; in the latter, SLE
also had a cold agglutinin of anti-i specificity that and lymphoma were the most frequently associated
reacted to a titer of 1024 at 4°C with cord cells and disorders. The patients tended to have severe hemo-
reacted with his own RBCs up to a temperature of lysis, which, although usually responding well to
33°C. Fractionation of the patient’s serum showed that treatment, ran a chronic course with intermittent exac-
the anti-i activity was confined to the IgM fraction. erbations, thus making overall management difficult.
Therapy with prednisone caused his warm autoanti- Shulman and coworkers217 reported that 12 of their
body to become weaker, but his hemoglobin level rose 144 patients (8.3%) with AIHA satisfied diagnostic crite-
only slightly, and there was a progressive rise in his ria for both WAIHA and CAS. All patients had cold
anti-i titer. He was therefore treated with cyclopho- agglutinins that reacted up to 37°C, although in none of
sphamide with marked improvement, which was the patients was the cold agglutinin titer at 4°C greater
maintained with the daily administration of pred- than 64. All 12 patients had severe hemolytic anemia
nisone, chlorambucil, and fluoxymesterone. that responded dramatically to corticosteroid therapy,
Moake and Schultz215 reported a 37-year-old with the mean hemoglobin level increasing from 6.3 to
woman with extravascular hemolytic anemia who 12.9 g/dL. Five patients had SLE, one patient had a non-
had positive antiglobulin and anticomplement DATs. Hodgkin’s lymphoma, and six patients had idiopathic
They demonstrated the presence of cold-reactive IgM, AIHA; four patients had concomitant thrombocytope-
IgA, and IgG antibodies, 37°C–reactive IgG antibod- nia. Shulman and coworkers suggested that such
ies, and depressed serum complement activity. patients represent a distinct category of AIHA charac-
In 1980, we reported four patients with AIHA who terized by severe hemolysis, typical serologic findings,
had serologic findings characteristic of both warm anti- and a dramatic initial clinical response to corticosteroid
body AIHA and CAS.1 The findings in these patients are therapy. The authors suggested that the patients should
summarized in Table 3-9. In all patients, the RBCs were be given corticosteroid therapy as soon as possible.
coated with IgG and C3. The cold agglutinin titers at Similar to the patients reported by Sokol and cowork-
4°C were 128, 1024, 2048, and 4096, and all of the anti- ers,216 most of the patients reported by Shulman and
bodies reacted up to 37°C. In three patients in whom the coworkers217 were found to develop chronic hemolysis.
cold agglutinin specificity was identified, the specificity Only in two patients was corticosteroid therapy tapered
was anti-i. All patients had a warm autoantibody off completely without a relapse of AIHA. Of the seven
80
TABLE 3-9. SUMMARY OF FINDINGS OF FOUR PATIENTS WITH AUTOIMMUNE HEMOLYTIC ANEMIA (AIHA) WHO HAD
SEROLOGIC FINDINGS CHARACTERISTIC OF BOTH WARM ANTIBODY AIHA AND COLD AGGLUTININ SYNDROME
DAT* IgG Warm Autoantibody
(Titer/Score) IAT* at Specificity
Reticulocytes 37°C with (routine
Patient Age (yr)/Sex Hb (g/dL) (%) Anti-IgG Anti-IgA Anti-IgM Anti-C3 Anti-IgG panel by IAT)

1 43/F 5.0 21 4096/47 0 0 16/15 Positive Not obvious


2 52/M 10.0 2.1 1024/35 0 0 32/26 Positive Not obvious
3 80/F 8.7 7.0 32,000/61 80/11 0 256/36 Positive Not obvious
4 28/F 4.5 12 512/28 20/6 40/16 32/30 Positive Not obvious
Cold Agglutinin Serum Serum Serum
4°C IgM IgG IgA
Patient (Titer/Score) Thermal Range Specificity (n = 45–200) (n = 664–825) (n = 59–311) Miscellaneous

1 4096/110 37°C Anti-i NT NT NT 1: Biclonal “M” spikes on serum


2 1024/94 37°C Anti-i 600 1166 83 electrophoresis; cryoglobulin
3 2048/102 37°C Unidentified 650 600 84 present.
4 128/59 37°C Anti-i NT NT NT
2: Further testing revealed anti-dl
and anti-Ena warm
autoantibodies.

Patient Associated Diagnosis Therapy Response

1 Lymphoproliferative disease Prednisone, cytotoxic drugs Hb improved to 9–10 g/dL; stable on therapy for 2 years.
2 Lymphoma Chlorambucil Hb Improved to 13.2 g/dL during 5-month follow-up.
3 Lymphoproliferative disorder Prednisone, cytotoxic drugs, Improved during short period of follow-up; Hb, 9.8 g/dL; reticulocytes,
splenectomy 2.4%
4 None Prednisone, splenectomy Gradual resolution of hemolysis; 2 years later, RBCs were weakly
coated with C3 only; IgG antibody no longer detected; IgM
antibody reactive only at 4°C.

* DAT, direct antiglobulin test; IAT, indirect antiglobulin test; NT, not tested.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 81

patients followed up who had chronic hemolysis, five Vick and coworkers224 described a patient with
showed persistence of both IgG warm autoantibodies chronic lymphocytic leukemia/small cell lymphoma
and high-thermal amplitude cold autohemagglutinins, who developed mixed type AIHA 3 weeks after her
whereas two lost their cold autohemagglutinins and fifth cycle of fludarabine. Prompt treatment with cor-
showed serologic findings indistinguishable from those ticosteroids and minimal blood transfusions led to
of warm antibody AIHA. marked improvement.
Nusbaum and Khosla218 reported a patient with
AIHA and a positive DAT for IgG. They believed that CAUTIONS CONCERNING THE DIAGNOSIS
the patient had combined cold and warm antibody OF COLD AND WARM AIHA
AIHA. However, no information was provided about
the DAT results using anticomplement reagents or the The diagnosis of mixed cold and warm antibody
thermal amplitude of the patient’s cold agglutinin. AIHA is sometimes made on the basis of inadequate
Without this information, it is impossible to ascertain serologic studies.218,221 As emphasized in Chapters 5
whether their patient had combined cold and warm and 6, appropriate characterization of the serum anti-
AIHA or warm antibody AIHA. bodies must be made before determining the specific
Freedman and coworkers30 reported a patient with type of AIHA present. An important source of poten-
AIHA who had three concomitant red cell autoanti- tial error is suggested by our data,1 which indicate
bodies: a low-titer, high-thermal amplitude IgM anti-I that 35% of patients with WAIHA have cold agglu-
cold agglutinin; an IgG warm panagglutinating auto- tinins reactive at 20°C. However, a large majority of
antibody; and an IgM warm hemolysin. The cold these cold agglutinins are clinically insignificant, and
agglutinin reacted at 4°C to a titer of 64 in saline and we found that only 5% reacted at 37°C. Unless one can
256 in albumin, and reacted up to 37°C. In addition, a document a cold autoantibody with a high thermal
strong IgG complement-binding platelet antibody was amplitude (>30°C) in association with a warm autoan-
demonstrated. The patient also had thrombophlebitis tibody, a diagnosis of cold and warm (“mixed”) AIHA
and developed gangrene of his right foot and lower leg, is not warranted.
requiring an above-knee amputation. After the admin- Some reports of cold and WAIHA do, indeed,
istration of prednisone, the patient’s hemolysis describe patients with serologic findings of warm
resolved and there was no recurrence of hemolysis. AIHA in association with classic serologic findings of
Kajii and coworkers219 reported that 3 of 46 CAS, that is, a high titer cold autoantibody with a
patients (6.5%) with AIHA satisfied the diagnostic high thermal amplitude.1,30,214,215,219 However, other
criteria for mixed-type AIHA. All of the cold auto- reports describe WAIHA plus cold autoantibodies
antibodies were IgM-κ and showed high titer and with normal cold agglutinin titers, but of high (30°C
high thermal amplitude. to 37°C) thermal amplitude217,225 (and we have per-
sonally observed numerous such cases). Finally,
SECONDARY COLD AND WARM AIHA some authors report “mixed” AIHA but do not
characterize the cold autoantibody other than stating
De Angelis and coworkers220 described a young HIV- that it was reactive in the cold and at warm
infected patient who presented with a severe AIHA temperatures.216,226
with both warm and cold autoantibodies. Serologic In Chapter 6, we review our experience with the
findings indicated the presence of a low-titer, high diagnosis of “cold and warm” AIHA and discuss the
thermal amplitude anti-I cold-reacting antibody and a appropriate extent of serologic testing.
pan-reactive warm autoantibody.
Similarly, Koduri and coworkers221 described three CLINICAL COURSE
acutely ill men with acquired immune deficiency
syndrome (AIDS) who presented with fever, spleno- Some authors suggest that patients with cold and
megaly, and severe AIHA with both warm and cold warm AIHA have a more severe onset and more
autoantibodies. However, no information was pro- chronic course than patients with other categories of
vided by the authors regarding the thermal amplitude AIHA. However, a comprehensive comparison with
of the cold agglutinins, so it is uncertain that these the clinical course of patients with warm or cold
were clinically important cold antibodies. AIHAs has not been made.
Chen and coworkers222 reported a 49-year-old
woman with primary biliary cirrhosis, sicca syndrome, MANAGEMENT
and mixed-type AIHA. She presented with life-threat-
ening anemia and was successfully treated with blood Initial therapy of mixed cold and warm AIHA is
transfusions and pulse methylprednisolone therapy. corticosteroids with additional measures as may
Gologan and coworkers223 reported a patient with become necessary depending on the patient’s course
Waldenström’s macroglobulinemia who also had (see Chapter 11). Morselli and coworkers227 described
WAIHA. During a period of compensated hemolysis the successful use of rituximab in a patient with
after treatment, there appeared a severe hemolytic mixed AIHA associated with an overt malignant
attack induced by transitory cold agglutinins with lymphoproliferative disease who relapsed after a
high thermal amplitude. course of corticosteroids.
82 Immune Hemolytic Anemias

AIHA Associated with a Negative DAT ative colitis and AIHA should be considered as only
two parts of a complex, immunologically mediated
This topic is reviewed in Chapter 9. multisystem disease state.2 In general, the evidence
for a relationship between immunologic disorders,
Secondary Autoimmune including immune deficiency states, and AIHA is
Hemolytic Anemias strong,2,236-242 although the pathogenetic basis for
such a relationship has not been clarified.
AIHAs are classified as secondary for any of several The significance of the relationship between other
reasons. One reason is the association of AIHA with associated diseases and AIHA is far less clear in many
an underlying disease with a frequency greater than reported cases of “secondary” AlHA. Occasionally
can be explained by chance alone. For example, all reported associated diseases include leukemias other
authors agree that the incidence of WAIHA is higher than CLL, thyroid disease, myeloproliferative disor-
in patients with CLL and SLE than in the general ders, pernicious anemia, cirrhosis of the liver, and
population. many bacterial infections. Hemolysis in many cases of
Another criterion for categorizing a given case of secondary hemolytic anemia is not caused by an
AIHA as secondary is the reversal of the hemolytic immune mechanism. Dacie243 has reviewed both
anemia simultaneously with the correction of the asso- immune and nonimmune secondary hemolytic
ciated disease. Ovarian tumors are a good example; anemias and provides many case reports, including
well-documented cases of cure of the AIHA after surgi- those in the older medical literature.
cal removal of the tumor have been reported.228-231
Still another reason for suspecting a relationship
between the occurrence of AIHA and an associated RELATIVE INCIDENCE OF IDIOPATHIC AND
disease consists of evidence of immunologic aber- SECONDARY TYPES OF WARM ANTIBODY AIHA
ration as part of the underlying disorder, especially if
the associated disease is thought to have an autoim- Many reports of the relative incidence of idiopathic and
mune pathogenesis. However, when the coexistence secondary forms of AIHA are available. The reported
of two immunologic disorders in a single patient is incidence varies significantly, possibly depending on
infrequent, the pathogenetic significance is conjectural the nature of the center to which cases were referred
and interpretations vary. For example, rheumatoid and also in regard to an interpretation of what consti-
arthritis is an extremely common disorder and reports tutes a related underlying disorder. Reports suggesting
of an associated AIHA are infrequent, so the combina- that idiopathic cases are more frequent include those of
tion may well be coincidental.232 Allgood and Dacie and Worlledge,5 Dausset and Colombani,10 and
Chaplin9 noted the presence of ulcerative colitis and Sokol and coworkers.4 An approximately equal inci-
rheumatoid arthritis in some of their patients but still dence is suggested by the reviews of Dameshek and
considered the AIHA idiopathic, whereas Pirofsky2 Komninos,244 Evans and Weiser,245 Bell and cowork-
included these and numerous other disorders among ers,189 and Engelfriet and coworkers.6 Reports suggest-
patients classified as having secondary AIHA. In ing that secondary cases are more common are those of
ulcerative colitis, reports of resolution of the Chaplin and Avioli246 and Pirofsky.2,8 Pirofsky’s data
hemolytic anemia after colectomy,233-235 the associa- are heavily influenced by the fact that his patient
tion of ulcerative colitis with other immunologically population was characterized by many patients with
mediated abnormalities, and the frequent sequential leukemia and lymphoma who were undergoing
development of immunologic abnormalities in a constant medical follow-up. Table 3-10 summarizes
single patient lend weight to the suggestion that ulcer- data available in these cited series.

TABLE 3-10. COMPARATIVE INCIDENCE OF IDIOPATHIC AND


SECONDARY WARM ANTIBODY AUTOIMMUNE HEMOLYTIC ANEMIA

No. of Idiopathic Secondary


Reference Cases No. % No. %

Dameshek and Komninos244 43 21 50 22 50


Dacie and Worlledge5 199 111 56 88 44
Dausset and Colombani10 106 83 78 23 22
Evans and Weiser245 37 15 41 22 59
Bell et al.189 37 18 49 19 51
Pirofsky2,8 234 44 19 190 81
Engelfriet et al.6 539 245 45 294 55
Sokol et al.4 (excluding drug induced) 1694 1034 61 660 39
Total 2889 1571 54 1318 46
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 83

TABLE 3-11. CLASSIFICATION OF 1834 PATIENTS WITH AUTOIMMUNE HEMOLYTIC


ANEMIA (AIHA), BASED ON SEROLOGIC FINDINGS AND DISEASE ASSOCIATION

Condition WAIHA CAS PCH Mixed AIHA

Idiopathic AIHA 617 344 11 82


Drug-Related AIHA 140 0 0 0

Neoplasia

Lymphoid neoplasms
Chronic lymphocytic leukemia 63 13 0 0
Acute lymphoblastic leukemia 1 4 0 0
Non-Hodgkin’s lymphoma 32 22 0 8
Angloimmunoblastic lymphadenopathy 1 1 0 0
Myeloma 4 1 0 0
Macroglobulinemia 2 3 0 0
Thyinoma 1 0 0 0
Hodgkin’s disease 10 4 0 4
Ovarian tumours 4 0 0 0
Carcinoma (nonovarian) 67 23 0 6
Acute nonlymphoid leukemias 5 4 0 0
Chronic myeloid leukemia 5 1 0 1
Myelofibrosis 12 0 0 3
Myelodysplastic syndromes 23 4 0 3
Other neoplasms 3 2 0 0

Infection

Pneumonia—Mycoplasma 0 29 0 1
Pneumonia—“viral” and unspecified 2 13 0 1
Infectious mononucleosis 0 6 0 0
Infectious hepatitis 1 1 0 0
Tuberculosis 0 1 0 0
Syphilis 0 0 1 0
Infection—unspecified 13 11 7 3

Collagen Diseases

Systemic lupus erythematosus 17 8 0 3


Rheumaroid arthritis 34 9 0 3
Polyarteritis nodosa 1 0 0 0

Other Immune-Based and Miscellaneous Disorders

Ulcerative colitis 19 1 0 1
Thyrotoxicosis 3 1 0 1
Myxedema 1 0 0 0
Chronic active hepaticis 2 0 0 0
Pernicious anemia 7 1 0 0
Diabetes mellitus 8 3 0 0
Sarcoidosis 1 0 0 0
Myasthenia gravis 1 0 0 0
AIHA Associated with Pregnancy 22 15 0 1
AIHA Associated with Chronic Renal Failure 29 12 0 6

Modified from Sokol RJ, Booker DJ, Stamps R: The pathology of autoimmune haemolytic anaemia. J Clin Pathol 1992;
45:1047–1052.

The data of Sokol and coworkers,4 which indicate OVARIAN TUMORS


the classification of 1834 patients with autoimmune
hemolysis, are indicated in Table 3-11. The authors Hemolytic anemia associated with ovarian tumors has
stressed that their series included patients who had been recognized since the report of West-Watson and
hemolysis but who were not anemic. Most of the Young in 1938.247 Their patient had hemolytic anemia
patients had warm autoantibodies (Table 3-12). and failed to improve after splenectomy. In an attempt
The following sections provide information about to find a hypothetical accessory spleen, the patient
secondary AIHA found in association with specific underwent further abdominal surgery, at which time
disorders. Further details about management of an ovarian dermoid cyst was discovered and excised.
secondary AIHAs are found in Chapter 11. Excision of the cyst was promptly followed by
84 Immune Hemolytic Anemias

ing specificity including anti-e, anti-c anti-E, and


TABLE 3-12. SEROLOGIC DISTRIBUTION
anti-dl. In three patients an autoantibody of anti-I
OF 1834 PATIENTS WITH AUTOIMMUNE
specificity was reported.
HEMOLYTIC ANEMIA
At least three hypotheses have been put forward to
explain the association of AIHA and ovarian tumors:
Autoantibody Type No. (%)
(1) the presence in the tumor of substances foreign to
Warm 1151 (62.8) the host that stimulate development of cross-reacting
Cold antibodies, (2) the binding of substances secreted by
Cold agglutinin disease 537 (29.3) the tumor to the RBCs with production of antibodies
Paroxysmal cold hemoglobinuria 19 (1.0) reactive with the coated RBCs, and (3) the tumor itself
Mixed (warm plus cold) 127 (6.9)
could produce red cell antibody. The latter hypothesis
Modified from Sokol RJ, Booker DJ, Stamps R: The pathology of autoimmune is supported by De Bruyere and coworkers,230 who
haemolytic anaemia. J Clin Pathol 1992;45:1047–1052. found that ovarian cyst fluid from a patient with
AIHA contained IgG antibodies of the same specificity
as in the patient’s serum. Similar findings have been
remission of the anemia. Particular notice was taken of reported by other authors.231,274
this result, and in subsequent years numerous case In conclusion, the association of AIHA with ovarian
reports230,248,249-251 and reviews228,229,231,252 of AIHA in tumors is rare but its recognition is obviously impor-
association with ovarian tumors have been published. tant. The possibility should be considered in all
Carreras Vescio and coworkers252 reviewed 29 women who develop AIHA of unknown origin, par-
published cases in 1983 and added one case of their ticularly if the AIHA is not responsive to standard
own228-231,247,248,252-274 (Table 3-13). Although most of therapeutic measures.
the patients studied were adults (average age,
40 years), the case reported by Allibone and Collins257 ULCERATIVE COLITIS
was of a girl only 4 years old. Twenty of the 30 ovarian
tumors were teratomas, including 17 dermoid cysts. The association of AIHA with ulcerative colitis was
In three cases, the tumors were reported as cysts, not first reported in 1955 by Lorber and coworkers.278
further described; in one patient, a probable theca-cell Although the association of the two disorders is quite
tumor was diagnosed, and the remaining patients had uncommon, there is undoubtedly a pathogenetic rela-
malignant neoplasms. In addition, the authors men- tionship because the AIHA has almost invariably gone
tioned that two cases of dermoid tumors in organs into remission after colectomy, even when hemolysis
other than the ovary have been associated with AIHA: is refractory to other therapeutic approaches.233,235,279
one was localized in the hilus of the spleen275 and the The incidence of AIHA in patients with ulcerative
other one in the mesentery.276 A further case of AIHA colitis in various reports has varied between 0.6%
and dermoid cyst of the mesentery was reported by and 1.7%.234,280-282 A positive DAT without hemolysis
Buonanno and coworkers.277 is found in an additional 2% of patients.282 Also, one
An important feature of AIHA in association with must be aware that patients taking sulfasalazine as
ovarian tumors is the striking resistance of the AIHA therapy for ulcerative colitis have developed
to any therapeutic approach other than the surgical hemolytic anemia,283 although such cases do not have
removal of the tumor. In 16 of the cases in the report an immune basis.284,285
of Carreras Vescio and coworkers,252 the hemolytic Dacie243 provided a detailed summary of cases of
anemia was first treated with steroids, which pro- AIHA in patients with ulcerative colitis that were
duced only a slight and transient improvement in six published up to 1979 (Table 3-14), and, in 1994,
instances. In seven patients, splenectomy was per- Ramakrishna and Manoharan235 reported two patients,
formed before excision of the ovarian tumor, but only and summarized data on 38 cases previously des-
three patients derived some benefit. In three instances, cribed in the English literature (Table 3-15). The treat-
excision of the ovarian tumor was carried out together ment and outcome and DAT status after therapy
with splenectomy; in all three cases surgery was fol- are summarized in Table 3-16. The therapeutic
lowed by complete recovery. In all of the cases, the approaches that did not include colectomy resulted in
ovarian tumor was eventually excised, and this was a remission of AIHA in about 50% of patients. In con-
followed by a generally total and rapid remission of trast, all 16 patients who had splenectomy combined
the AIHA, except that only partial benefit occurred in with colectomy or colectomy alone went into remis-
three patients who had malignant tumors with sion of their AIHA. Patients who have developed
metastatic disease. In one patient who had a malig- AIHA many years after undergoing total colectomy
nant tumor with metastatic disease, complete remis- for severe ulcerative colitis refractory to medical man-
sion of the anemia did occur after removal of the agement have been successfully treated with cortico-
tumor and chemotherapy. steroids and/or immunomodulators.281,286
The DAT was positive in 22 cases, negative in only There is general agreement that the initial therapy
3; data were not reported in the remaining 5 patients. for AIHA in association with ulcerative colitis should
Most patients had IgG autoantibodies, some exhibit- be steroid therapy, as it has been successful in about
TABLE 3-13. REPORTED CASES OF HEMOLYTIC ANEMIA ASSOCIATED WITH OVARIAN TUMORS

Age Duration Palpable Hemoglobin Reticulocytes Coombs Tumor Histologic


Patient (yr) of Illness Splenomegaly Tumor (q/100 mL) (%) Test Corticosteroids Splenectomy Excision Diagnosis

1 44 2 mo + + 3.6 45 + — No response Remission Teratoma


2 19 2 yr + + 5.0 15 + — At time of Remission
tumor “Cyst”
excision
3 47 5 mo + + 5.1 46 ? — Transient Remission Dermoid
response
4 35 1 mo ? + 5.0 10 + — — Slow Pseudomucinous
response cystadeno-
carcinoma
5 40 ? + + 4.3 16 + — No response Died at Dermoid
operation
6 4 3 mo ? + 7.0 39 0 — — Remission Dermoid
7 61 9 mo 0 0 5.2 20 ? — — Died of Dermoid
transfusion
8 53 ? 0 ? 6.5 78 + No response — Remission Dermoid
9 54 ? 0 + PCV: 23% 58 + No response — Remission Dermoid
10 40 ? + ? 4.3 74 + Transient Transient Remission Dermoid
response response
11 26 1 yr + + 3.6 26 + — — Remission Teratoma
12 44 9 mo + + 4.0 50 + No response No response Remission Dermoid
13 61 2 yr + + PCV: 37% 39 + No response — Remission Teratoma
14 23 1 yr 0 + 6.8 11 + — — Remission “Cyst”
15 39 4 mo ? ? ? ? + — — Remission Cystic
degeneration
16 49 ? ? ? ? ? ? No response No response Remission Dermoid
17 45 ? ? + ? ? + — — Remission Dermoid
18 30 4 mo + + 3.5 74 + Partial — Remission Dermoid
response
19 28 ? + + 6.7 5.7 ? Partial — Partial Papillary
response response adenocarcinoma
20 48 3 mo + + 4.2 9.7 0 No response — Remission Dermoid
21 36 1 yr + + PCV: 14% 8.6 + Partial — Incomplete Anaplastic
response carcinoma
22 52 9 mo 0 0 6.8 11 + No response At time of Remission Dermoid
tumor
excision
23 54 ? ? ? ? ? ? ? ? Partial Adenocarcinoma
response
24 64 1 yr 0 + 4.75 10.2 + No response — Transient “Epithelio-
response sarcomatoid”
tumor
25 41 1 yr + 0 3.8 4.2 0 — At time of Remission Theca-granulosa
tumor
excision
26 32 8 mo + + 3.5 50 + No response Transient Remission Dermoid
response
27 35 1 mo + + 5.5 6.5 + — — Remission Dermoid
28 47 4 mo 0 + 4.8 60 + Partial — Remission Dermoid
response
29 34 1 mo 0 + 10.3 1.9 + Partial — Remission Dermoid
response
30 29 3 mo 0 + 7.3 5.2 + No response — Remission Anaplastic
carcinoma

85
Modified from Carreras Vescio LA, Toblli JE, Rey JA, Assaf ME, Deinaria HE, Marletta J: Autoimmune hemolytic anemia associated with an ovarian neoplasm. Mediciria (B Aires) 1983;43:415–424.
86 Immune Hemolytic Anemias

TABLE 3-14. CASE REPORTS IN CHRONOLOGIC ORDER OF AUTOIMMUNE HEMOLYTIC ANEMIA (AIHA)
ASSOCIATED WITH ULCERATIVE COLITIS (UC)

Treatment and
Time of Onset of Maximum Serologic Findings: Response to
Age (yr)/Sex AIHA after Onset of Minimum Reticulocyte DAT; Specificity Treatment;
Patient of Patient UC Hb (g/dL) Count (%) of Autoantibody Other Findings

1 28/F 5 mo 8.2 10.4 DAT 3+. Anti-e; also Partial response


allo–anti-D and to cortisone
allo–anti-E and a and ACTH;
panantibody further
improvement
followed subtotal
colectomy. Blood
loss had
contributed to her
anemia
2 44/F 19 mo 9.0 2 DAT 1+. Panantibody ACTH controlled
active against intestinal bleeding.
enzyme-treated cells Probably no
significant
hemolysis
3 44/F 21 yr 6.0 72 DAT 4+. Panantibody; Splenectomy led to a
also allo–anti-E good but
incomplete
remission. DAT 1+
5 yr later
4 17/F 6 mo 10.8 0.8 DAT weak (±) pos. No treatment needed
Panantibody; also for mild anemia,
allo–anti-E and except transfusion
allo–anti-K before colectomy

5 31/F DAT pos. Anti-e + f Patient had had


eluted rheumatic heart
disease. DAT neg
6 weeks after
colectomy
6 35/F 6 yr 9.0 6.0 DAT 4+. Antibody 51Cr T
50 9 days. A
7S γG; no specificity high incidence of
demonstrated gastrointestinal
disorders and
allergies in
patient’s family,
including
pernicious anemia
7 23/M 6 mo 4.0 — DAT and IAT pos Splenectomy; after
postoperative
complications, a
normal blood
picture
8 21/M 1 yr Hematocrit — DAT pos Rheumatoid arthritis
18% 1 year before UC.
Splenectomy
(spleen weighed
1850 g); good
response
9 10.3 1.8 DAT 3+ RBC survival normal
10 57/M 11 yr after severe 10.2 9.7 DAT strong pos Remission after
UC treated prednisone
succesfully by therapy,
colectomy and although DAT
protectomy remained pos
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 87

TABLE 3-14. CASE REPORTS IN CHRONOLOGIC ORDER OF AUTOIMMUNE HEMOLYTIC ANEMIA (AIHA)
ASSOCIATED WITH ULCERATIVE COLITIS (UC)—CONT’D

Treatment and
Time of Onset of Maximum Serologic Findings: Response to
Age (yr)/Sex AIHA after Onset of Minimum Reticulocyte DAT; Specificity Treatment;
Patient of Patient UC Hb (g/dL) Count (%) of Autoantibody Other Findings

11 23/F 2 yr 4.4 52 DAT pos. IgG; Remission after


antibody prednisone therapy
predominantly anti-e
12 18–58/3M, 3–225 mo Hematocrit 6.3 DAT pos Four patients died
2F 16.1% 2–7 mo after onset
of pos DAT.
13 42/F 1 yr 5.0 18 DAT strong pos Hemolysis failed to
respond to medical
treatment. Rapid
emission followed
total colectomy
14 32/M 3 yr after 2.58 × 22.5 DAT pos. IgG; anti-e Complete recovery
colectomy for 1012/L from hemolysis
UC RBCs after proctectomy
15 23/F Known case of UC 9.8 7 DAT pos. IgG; anti-e Hemolysis responded
to prednisone
therapy
16 30/F 1 yr after onset of 7.8 80 DAT pos. Anti-Rh Hemolysis responded
mild UC to splenectomy
17 36/F Shortly after onset 8.0 11.5 DAT pos. IgG; anti-e Large doses of
or anti-C corticosteroids and
6MP needed to
control hemolysis;
remitted after 5 yr;
UC also quiescent
18 35/F 10 yr 11.0 2 DAT and IAT strong DAT became neg 4
pos days after total
colectomy
19 34/F 7 yr Hematocrit 44 DAT 4+. IgG; Incomplete response
21% anti-Rh (neg of hemolysis to
with Rhnull cells) prednisone;
remission followed
splenectomy
20 12/M 1 mo Hematocrit 7.8 DAT 4+. Nonspecific Incomplete response
26% of hemolysis to
prednisone;
remission followed
splenectomy. Total
colectomy when
aged 23; 1 yr
later hematocrit
51%
21 24/F Simultaneous onset Hematocrit 5 DAT 4+. IgG; Incomplete response
of UC and AIHA 25.5% anti-Rh (neg with of hemolysis to
Rhnull cells) prednisone and
cyclophosphamide.
Splenectomy
ineffective.
Hematocrit
stabilized at at
35% after
coloproctectomy
88 Immune Hemolytic Anemias

TABLE 3-14. CASE REPORTS IN CHRONOLOGIC ORDER OF AUTOIMMUNE HEMOLYTIC ANEMIA (AIHA)
ASSOCIATED WITH ULCERATIVE COLITIS (UC)—CONT’D

Treatment and
Time of Onset of Maximum Serologic Findings: Response to
Age (yr)/Sex AIHA after Onset of Minimum Reticulocyte DAT; Specificity Treatment;
Patient of Patient UC Hb (g/dL) Count (%) of Autoantibody Other Findings

22 29/F 5 yr 7.2 24 DAT pos Incomplete response


of hemolysis to
prednisone;
improved after
splenectomy and
azathioprine
23 30/F 4.5 yr 9.3 20 DAT pos Total proctocolectomy
and splenectomy.
2.5 yr later no
evidence of
hemolysis
24 23/M 1.5 yr Hematocrit 52 DAT 4 + Incomplete response
15.6% to prednisone.
Splenectomy; 4 mo
later no evidence
of hemolysis

Modified from Dacie JV: The Haemolytic Anaemias, 3rd ed. vol. 4, Secondary or Symptomatic Haemolytic Anemias. New York: Churchill Livingstone, 1995.

half of the cases.234,287 Several authors suggest that ation of RBC antigen in the gastrointestinal tract result-
patients unresponsive to steroids should undergo ing in the production of antibodies against the altered
total proctocolectomy directly, even in the absence of antigens, which may react with the patient’s native
active colonic disease.283,288 Other authors recom- RBCs; (2) absorption of non-RBC antigens through the
mend splenectomy before total colectomy.234,281 diseased colon with the development of antibodies
Proctocolectomy has the added advantage of remov- that cross-react with the patient’s RBCs; (3) interaction
ing the risk of primary colonic cancer in longstanding of anticolon antibodies with RBC antigens; and
ulcerative colitis289 and avoids the risks of splenec- (4) nonspecific stimulation of the immune system with
tomy (see Chapter 11). the appearance of a clone of immunocompetent cells
Mechanism of AIHA. The mechanism of the AIHA producing anti-RBC antibodies.233,290,291
in patients with ulcerative colitis remains obscure. Yates and coworkers292 studied the ability of
Proposed theories include the following287: (1) alter- mononuclear cells extracted from the colon, draining
lymph nodes, peripheral blood, and spleen of a patient
with severe AIHA to produce RBC autoantibodies.
TABLE 3-15. PATIENTS WITH AUTOIMMUNE They demonstrated that in vitro cultures of mononu-
HEMOLYTIC ANEMIA (AIHA) AND ULCERATIVE clear cells of the colon produced immunoglobulin
COLITIS (UC) spontaneously, showing the presence of activated B
cells and plasma cells, but anti−red cell activity could
Clinical Features Patients, n not be demonstrated. However, mononuclear cells
Sex Male 16
transferred to SCID mice did produce IgG with anti−
Female 24 red cell activity. The authors concluded that the colon is
Age range (yr) 12–40 28 the source of RBC autoantibodies in such patients.
41–60 12
Activity of colitis Active 36 LYMPHOPROLIFERATIVE DISORDERS
at the time Inactive 4
of AIHA episode AIHA followed colectomy 2 The records of 637 patients with lymphoproliferative
UC diagnosed after AIHA 2 disorders and 346 patients with myeloproliferative
DAT Positive 40
IgG only 15 disorders were retrospectively analyzed for the
IgG and complement 3 presence of coexistent autoimmune derangements.293
Complement only 1 The frequency of autoimmune perturbations in
Not specified 21 lymphoproliferative diseases (51 cases; 8.0%) was
Specificity Known 8
Anti-e 6
significantly higher than in myeloproliferative dis-
Anti-E 1 eases (6 cases; 1.7%; P < 0.0001). Rheumatic disorders,
Anti-C 1 autoallergic hematologic manifestations, and other
Unknown 32 organ-specific autoimmune derangements were res-
Modified from Ramakrishna R, Manoharan A: Auto-immune haemolytic
ponsible for about one third each of the observed dis-
anaemia in ulcerative colitis. Acta Haematol 1994;91:99–102. turbances. These data are similar to those of Miller,294
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 89

TABLE 3-16. PATIENTS WITH AUTOIMMUNE HEMOLYTIC


ANEMIA AND ULCERATIVE COLITIS

Total Outcome:
No. Grade of Response
Treatment of Patients I II III

Steroids 18 5 2 11
Steroids + immunosuppressives 16 7 3 6
Steroids + danazol 1 — 1 —
Splenectomy 8 1 3 4
Splenectomy + colectomy 6 6 — —
Colectomy 10 10 — —

Grade I, hemolysis and colitis in remission, and DAT either negative (n = 12) or weakly
positive (n = 8) or unknown (n = 9); grade II, hemolysis in remission, colitis activity unknown
or not responsive, DAT status unchanged: grade III, unresponsive or unknown hemolytic and
colitis status.
Modified from Ramakrishna R, Manoharan A: Auto-immune haemolytic anaemia in ulcerative
colitis. Acta Haematol 1994;91:99–102.

who observed diffuse connective tissue diseases and marizes six of the largest series evaluating the rela-
autoimmune hemolytic anemias in 6.8% of his lym- tionship of AIHA and malignancy.295,302 CLL is the
phoma patients. diagnosis in 15% of all warm antibody AIHA patients
Autoimmune diseases that preceded the onset of and half of the malignant cases, and is exceeded only
malignancy occurred in lymphoproliferative and by idiopathic cases. In contrast, these data indicate
myeloproliferative disorders with a comparable fre- that CAS is only rarely associated with CLL. However,
quency without significant differences between indi- later reports regarding the incidence of AIHA in
vidual subgroups of lymphoproliferative diseases. In patients with CLL indicated that hemolysis mediated
contrast, autoimmune complications developing in by IgM cold antibodies was found in 7 of 52 cases
the course of the neoplastic disease were signifi- (13%) by Mauro and coworkers296 and in 13 of 76
cantly more frequent in lymphoproliferative (4.9%) patients (17%) by Sokol and coworkers.4
than in myeloproliferative disorders (0.3%; P < 0.0005). When CLL is associated with AIHA, the CLL is
Here marked differences were observed between rarely occult. In collected series of more than
individual lymphoma entities, the rate of concomitant 500 patients, the AIHA antedated the diagnosis of
autoimmune derangements ranging from zero to CLL in a single patient.295 A higher lymphocyte count,
over 15%. older age, and male gender are significantly linked
Chronic Lymphocytic Leukemia. CLL is not only a with an increased rate of AIHA at CLL diagnosis.296
malignant disease but also a complex immunologic Also, therapeutic approaches, such as radiation and
disorder. The immune dysregulation that is a hall- alkylating agents, particularly purine analogues, have
mark of CLL manifests itself, on the one hand, in an been considered as risk factors for the occurrence of
immunodeficiency state and, on the other hand, in an AIHA. It is generally thought that fludarabine may
excess of autoimmune phenomena.295 predisopose to AIHA by inducing a marked lympho-
AIHA is the most common autoimmune disease cytopenia, particularly of CD4+ lymphocytes, with a
associated with CLL, and CLL is the most common of T-cell subset imbalance that may favor the emergence
the known causes of AIHA.6,295 Various reports of autoreactive T cells (see Chapter 8).
indicate that AIHA occurs in 5% to 37% of CLL The mean time from diagnosis of CLL to AIHA is
patients.295-301 The figures are highly dependent on 4.1 years; lymphadenopathy is present in 88%;
the duration and extent of disease.295 Table 3-17 sum- splenomegaly is present in 94%, and it is massive

TABLE 3-17. AUTOIMMUNE HEMOLYTIC ANEMIA (AIHA) AND MALIGNANCY

Total
Associated
Idiopathic with Malignancy CLL NHL HD WM/MM ALL/AML Other Infection
n (%) (%) (%) (%) (%) (%) (%) (%) (%)

Warm AIHA 1463 47 31 15 4 3 2 3 5 0


Cold AIHA 392 48 21 1 6 3 2 2 10 24

CLL, chronic lymphocytic leukemia; NHL, non-Hodgkin’s lymphoma; HD, Hodgkin’s disease; MM, multiple myeloma; WM, Waldenstrom’s macroglobulinemia; ALL,
acute lymphocytic leukemia; AML, acute myeloid leukemia; AIHA, autoimmune hemolytic anemia.
From Diehl LF, Ketchum LH: Autoimmune disease and chronic lymphocytic leukemia: Autoimmune hemolytic anemia, pure red cell aplasia, and autoimmune
thrombocytopenia. Semin Oncol 1998;25:80–97.
90 Immune Hemolytic Anemias

in 20%. The peripheral blood smear demonstrates Pathophysiology. Although pathogenic RBC auto-
microspherocytes, polychromasia, and anisocytosis. antibodies in CLL patients with AIHA occasionally
Normoblasts are present in about one third of may be made by the clone of malignant B cells,309-312
patients, and there is even an occasional early myeloid they are generally produced by remnant normal
cell⎯all changes typical of stimulated erythropoiesis. B cells.295,296,309,313 The autoantibodies are generally
The peripheral blood film virtually always contains polyclonal, and the Ig isotypes of autoantibodies
the findings consistent with CLL.295,303 found in RBC eluates usually differ from that of the Ig
Serologic Findings. In the great majority of cases of expressed by the malignant B-cell clone.
CLL complicated by AIHA, the responsible autoanti- The development of antierythrocyte autoantibodies
bodies have been of the warm type and similar in class, in CLL patients may reflect a generalized defect in the
specificity, and behavior in vitro to those associated homeostatic mechanisms that prevent generation of
with idiopathic AIHA. Engelfriet and coworkers304 antiself humoral immunity.295,309 According to this
reported that of 130 patients with AIHA who had an hypothesis, autoantibodies arise not from polyclonal
underlying malignant disease, 79 had CLL and that all B-cell activation, but rather from a pathologic immune
had formed warm antibodies. The serologic findings in response to self-antigen.309
11 cases with warm AIHA and CLL reported by Barcellini and coworkers314 studied 69 B-CLL
Dacie243 are listed in Table 3-18. However, there have patients and 53 controls using a recently developed
been some reports of cold antibody AIHA in associa- mitogen-stimulated DAT (MS-DAT), which is able to
tion with CLL.243,305,306 Ruzickova and coworkers307 disclose latent anti-RBC autoimmunity in AIHA. They
reported a patient with a 7-year history of idiopathic investigated the prevalence of anti-RBC autoimmunity
CAS in whom B-CLL subsequently developed. They by MS-DAT and the pattern of cytokine production by
determined that the B-CLL had developed from the PHA-stimulated whole blood cultures. In B-CLL the
patient’s cold agglutinin−producing B-cell population. prevalence of anti-RBC autoimmunity was 28.9% by
Prognosis. Several studies have shown that CLL MS-DAT, compared with 4.3% by the standard DAT.
patients with AIHA represent a poor prognosis cate- Production of numerous cytokines was significantly
gory,297,301,308 whereas a more recent study by Mauro increased in all B-CLL patients compared with controls.
and coworkers296 found in a multivariate analysis that They concluded that their data were consistent with the
AIHA has no independent effect on survival probabil- hypothesis that autoimmune phenomena in B-CLL are
ity. However, two independent factors were associated with an imbalance toward a Th-2−like
significantly related to better survival probability of profile. The elevated prevalence of anti-RBC autoim-
CLL patients with AIHA: the IgG class of the autoan- munity found by MS-DAT suggests that an underesti-
tibody and the occurrence of AIHA at the time of CLL mated latent autoimmunity exists in B-CLL.
diagnosis. Patients with IgM autoantibody identified Hodgkin’s Disease. AIHA is an uncommon occur-
a small group with very poor survival. In these cases, rence in patients with Hodgkin’s disease. In 1967, Eisner
the IgM autoantibody was a warm agglutinin, opti- and coworkers315 reported 10 patients with Hodgkin’s
mally reactive at 37°C (Fig. 3-6). disease who had a positive DAT. Overt hemolytic
anemia was present in eight of the patients, and the inci-
1.0 dence of AIHA in a series of 219 patients was 2.7%. With
0.9 one exception, all of the patients had advanced disease
at the time of detection of the positive DAT.
0.8
Levine and coworkers316 reviewed 71 cases of
0.7 Hodgkin’s disease and found three who had AIHA
Probability

0.6 (4.2%). A positive DAT without hemolysis was found


0.5 in an additional 4 patients. These authors emphasized
IgG that all of their patients had active disease at the time
0.4
IgM
a positive DAT was first noted, although there have
0.3 been reports of AIHA in patients with limited stage
0.2 disease.317-319 In some patients the AIHA was diag-
0.1 p < 0.001 nosed 3 or more years before the diagnosis of
Hodgkin’s disease,320-324 whereas in other patients
0.0
0 12 24 36 48 60 72 84 96 AIHA has supervened after a long interval after the
Months diagnosis of Hodgkin’s disease.325-329
FIGURE 3-6. Actuarial survival probability of chronic lymphatic leukemia Xiros and coworkers330 found only 1 patient with
patients from autoimmune hemolysis diagnosis according to the Ig AIHA among 492 patients with Hodgkin’s disease
class of the autoantibody. Percent surviving at 2 years: IgG (45 (0.2%). Additional case reports of patients with AIHA
patients), 76% (SE, ±6.6); (7 patients), 21% (SE, ±17) (P < 0.001). in Hodgkin’s disease have appeared in the medical lit-
(From Mauro FR, Foa R, Cerretti R, Giannarelli D, Coluzzi S, Mandelli F,
et al.: Autoimmune hemolytic anemia in chronic lymphocytic leukemia:
erature,243,331-334 including cases in children.317,335-338
Clinical, therapeutic, and prognostic features. Blood 2000;95: Most reports indicated that the presence of AIHA does
2786–2792.) not signal a bad prognosis.316,339
TABLE 3-18. SEROLOGIC FINDINGS IN AUTOIMMUNE HEMOLYTIC ANEMIA ASSOCIATED WITH CHRONIC LYMPHOCYTIC LEUKEMIA

Lysis
Agglutination
Papainized
DAT IAT Normal RBCs Papainized Papainized Normal RBCs
(titer) Normal RBCs PNH PNH Specificity
Age (yr)/ Anti– (titer) RBCs RBCs Stored Fresh Type of of
Patient Sex Anti-γ G non-γ 37°C 2–4°C 20°C 37°C 37°C 37°C 37°C 37°C Autoantibody Autoantibody

1 71/M +++ — + ... <1 + — — — — Warm Anti-e + NS


2 57/M ++ — + <1 <1 64 — — — — Warm NS
3 73/F ++++ — + 16 <1 1024 — ++ + — Warm NS
4 77/M ++ — — <1 <1 <1 — — — — Warm NS
5 51/F +++ ++ — <1 <1 16 ... ... ... ... Warm ? NS
6 58/M ++ + + <1 <1 16 ... ... ... ... Warm NS
7 65/M +++ + ++ <1 <1 16 — — — — Warm NS
8 68/F +++ — — ... ... 1 — — — — Warm NS
9 72/M +++ — — ... <1 <1 — — — — Warm NS
10 53/M ++ — — <1 <1 <1 — — — — Warm ...
11 65/M — +± — ... <1 1 ... ... ± — Warm NS

++++ denotes strong naked-eye agglutination or haemolysis; +++, ++, +±, +, and ± denote lesser degrees of agglutination of hemolysis; . . . denotes no observation; NS denotes no specificity demonstrated.
Modified from Dacie JV: The Haemolytic Anaemias, 3rd ed. vol. 4, Secondary or Symptomatic Haemolytic Anaemias. New York: Churchill Livingstone, 1995.

91
92 Immune Hemolytic Anemias

Autoantibody Specificity. In almost all patients


with AIHA associated with Hodgkin’s disease, the TABLE 3-19. SUMMARY OF THE
autoantibody has been found to be warm in type and CHARACTERISTICS OF 16 PATIENTS WITH
not distinctly separable in respect of antibody class NON-HODGKIN’S LYMPHOMA AND AUTOIMMUNE
and specificity from the warm antibodies responsible HEMOLYTIC ANEMIA
for idiopathic AIHA.243 An exceptional case was
n (%)
reported by Brain and coworkers,323 who described a
patient who had had a cold-antibody AIHA for Age (yr)
approximately 4 years before recurrence of severe Median 68
anemia and enlarging lymph nodes led to the diagno- Range 33–82
sis of an anaplastic type of Hodgkin’s disease. The Sex
Male 4 (25)
autoantibody was a high thermal amplitude anti-I. Female 12 (75)
In 1972, Garratty and coworkers340 described the Stage of NHL
presence of an autoantibody with anti-IT specificity in II 4 (25)
an American patient with Hodgkin’s disease. The III 2 (12.5)
antibody in this patient was of the IgG class, was tran- IV 10 (62.5)
AIHA
sient in nature, and was not associated with overt Warm 13 (81)
hemolysis. In a later report, Garratty and coworkers328 Cold 3 (19)
reported three patients with Hodgkin’s disease who Serum Ig
had AIHA associated with an IgG auto-anti-IT. IgG 5 (31)
IgM 4 (25)
Antibody of this specificity was not found in 50 Median time of response for NHL (days) 65
patients with Hodgkin’s disease who had a negative Median time of response for AIHA (days) 9
DAT, nor was it found in a group of 70 patients with Follow-up (mos)
AIHA associated with a variety of underlying dis- Median 14.5
eases, including non-Hodgkin’s lymphoma. Range 3–38
However, not all patients with auto-anti-IT have From Sallah S, Sigoumas G, Vos P, Wan JY, Nguyen NP: Autoimmune
Hodgkin’s disease. Freedman and coworkers341 hemolytic anemia in patients with non-Hodgkin’s lymphoma: Characteristics
and significance. Ann Oncol 2000;11:1571–1577.
reported a patient who did not have Hodgkin’s disease
but who had AIHA due to an autoagglutinin of anti-IT
specificity. The antibody was of the IgM class, bound
complement, and reacted optimally at 37°C. Haffleigh Non-Hodgkin’s Lymphomas. The association
and coworkers342 described the presence of anti-IT between AIHA and non-Hodgkin’s lymphoma (NHL)
occurring in three patients who did not have Hodgkin’s has been described in both B- and T-cell disorders.
disease, although two had non-Hodgkin’s lymphoma. Sallah and coworkers345 found AIHA in 16 of 501
None of these patients had evidence of shortened red patients (3%) with NHL. (Patients with small lympho-
cell survival. Levine and coworkers316 also found one cytic lymphoma and angioimmunoblastic lym-
patient with anti-IT in whom overt hemolytic anemia phadenopathy with dysproteinemia were excluded
was not present, but reported two additional cases with from their analysis.) A similar prevalence was reported
anti-IT in patients who did have active hemolysis. by Gronbaek and coworkers.346 The occurrence of
AIHA caused by cold agglutinins of anti-IT AIHA was not statistically significant among the four
specificity have also been reported by Postoway and stages of NHL (P = 0.722).345 In comparison with the
coworkers343 and by Ramos and coworkers.344 In both cohort of 501 patients with NHL evaluated during the
cases the cold agglutinin reacted with cord (i) > adult study period, patients with NHL and AIHA were more
(I) > adult (i). The patient reported by Postoway and likely to have T-cell NHL (33% versus 14%; P = 0.047),
coworkers343 was a 73-year-old woman with non- as has also been reported by other investigators.347,348
Hodgkin’s lymphoma who developed hemolytic Sallah and coworkers described the clinical, labora-
anemia with a positive DAT caused by C3. A cold tory, and pathologic features of their 16 patients with
agglutinin was present that reacted to a titer of 256 NHL and AIHA (Table 3-19). They found that the diag-
against cord RBCs and was reactive up to 37°C. The nosis of AIHA preceded that of NHL in nine patients,
patient reported by Ramos and coworkers344 was a 54- while in seven patients AIHA occurred either at the
year-old man who had progressively severe cold time of diagnosis of NHL or during the course of the
AIHA for 9 months. The DAT was weakly positive for disease. Resolution of the AIHA was achieved in 14
IgM and C3d, and the serum gave a bi-thermic patients at a median time of only 9 days. However,
(“Donath-Landsteiner−like”) reaction and contained a AIHA subsequently recurred in six patients. In five of
cold agglutinin with wide thermal amplitude and these patients, the recurrence of AIHA coincided with
anti-ITP specificity, reacting to a titer of 640 against that of NHL, while in one patient, it reappeared
cord cells. The patient was treated aggressively but shortly after splenectomy and during hospitalization
died of sepsis during an unsuccessful trial of chlo- for sepsis. Both NHL and AIHA were refractory to
rambucil. Autopsy revealed disseminated non- chemotherapy in two of the three patients with cold
Hodgkin’s lymphoma. agglutinin disease. The patients with NHL who did
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 93

not develop AIHA had better overall survival and Development of Lymphoproliferative Disorders
median survival compared with the NHL/AIHA as a Complication of AIHA. Sallah and coworkers357
group (P = 0.006 and P = 0.0001, respectively) (Fig. 3-7) evaluated 107 patients with idiopathic or secondary
CAS in Non-Hodgkin’s Lymphoma. Although AIHA and found that 19 (18%) eventually developed
patients with CAS constitute the minority of cases of malignant lymphoproliferative disorders. The median
secondary AIHA in NHL, a number of reports have time to develop malignancy was 26.5 months (range,
emphasized this association.349-355 Isbister and 9 to 76 months). Using multivariate analysis,
coworkers356 described four patients with lympho- advanced age (P = 0.005), underlying autoimmune
proliferative disorders who developed severe CAS. In diseases (P = 0.002), and the presence of serum gam-
three of the patients, the cold antibody was in low titer mopathy (P = 0.45) were risk factors for future devel-
(8 to 64) at 4°C, but had a wide thermal range (30°C to opment of lymphoproliferative disorders in these
37°C); the fourth patient’s cold antibody was present patients. Also, serum monoclonal IgM protein was a
to a titer of 2084 at 4°C. In two patients, the antibody significant predictor (P = 0.0001). More than one third
was a panagglutinin, and in the other two, including of the patients in this study had underlying autoim-
the patient with a high titer antibody, the specificity mune diseases, and immunosuppression had been
was anti-I. Despite a fulminant hemolytic presenta- used on an intermittent or continuous basis to manage
tion in all four cases, resolution of hemolysis occurred immune phenomena in these patients. Thus, it is pos-
after splenectomy or corticosteroid therapy. sible that immunosuppressants potentiate the risk of
Economopoulos and coworkers351 reviewed 370 lymphoproliferative disorders in a subset of patients
patients with NHL and found that 23 (6.2%) had with AIHA and autoimmune disorders.
AIHA, 4 of whom (1.1%) had cold-reacting antibodies. A further case was reported by Jasty and cowork-
These cases were all associated with IgM-κ serum ers,358 who described a 9-month-old boy who had
paraproteins, and the specificity of the autoantibody Evans’s syndrome and was treated with IVIG and cor-
was anti-I in three of the four cases. All of the patients ticosteroids. The infant eventually had recurrent
had low-grade lymphomas with splenomegaly and fevers, hepatosplenomegaly, pulmonary nodules, and
bone marrow involvement, and all had profound parenchymal central nervous system lesions. Results
hemolytic anemia. Three of the four patients of a lung biopsy revealed a polyclonal lymphoprolif-
responded to glucocorticoids and alkylating agents, erative disease, and polymerase chain reaction analy-
whereas the fourth patient responded poorly and died sis showed the presence of the EBV genome in the
with generalized lymphoma and active AIHA. lung nodules. The infant died from progressive lung
Pascali352 observed CAS in 3 of 62 (4.8%) patients with disease 6 months after the initial symptoms of Evans’s
NHL. Monoclonal IgM-λ proteins were present in two syndrome.
patients and IgM-κ in the other. Chaplin359 described two patients who had had
Also, see Waldenström’s Macroglobulinemia (page CAS for 10 and 7 1⁄2 years, respectively, before devel-
113). oping malignant lymphomas. Both patients had

1.00

0.75
Survival distribution function

FIGURE 3-7. Kaplan-Meier estimate of survival for the


patients with non-Hodgkin’s lymphoma/acquired immune
hemolytic anemia (interrupted line) and the cohort (straight
line). The difference between the two groups was 0.50
statistically significant (log-rank test; P < 0.0001). (From
Sallah S, Sigounas G, Vos P, Wan JY, Nguyen NP:
Autoimmune hemolytic anemia in patients with non-
Hodgkin’s lymphoma: Characteristics and significance. Ann
Oncol 2000;11:1571–1577.)
0.25

0.00
0 20 40 60 80 100 120
Time to follow-up (months)
94 Immune Hemolytic Anemias

received chlorambucil as treatment for their hemolytic associated with warm autoantibodies, Nair and
anemia, with the cumulative doses being 14.0 and coworkers375 reported a woman with CAS that devel-
6.6 g. Otto and coworkers360 described the develop- oped during the course of SLE. The cold antibody titer
ment, in a 66-year-old man who had had CAS of IgM- was 4096 and the autoantibody had anti-I specificity.
κ type for about 11 years, of increasing numbers of A low level of serum complement is commonly
malignant cells in his bone marrow. The findings were found in SLE. As shown in Table 3-20, the level was
interpreted as indicating a slow but progressive low or very low in six out of seven cases tested among
change toward malignancy. Kamiyama and cowork- a series of patients reported by Dacie.243
ers361 described two patients who developed NHL 7 1⁄2 It is not certain whether AIHA portends a more
and 8 years, respectively, after having initially pre- benign or more severe prognosis for patients with
sented with AIHA. SLE. Two studies found that AIHA was associated
with increased mortality,367,376 but AIHA was not an
SYSTEMIC LUPUS ERYTHEMATOSUS independent predictor of mortality once other mani-
festations (e.g., thrombocytopenia and renal disease)
Approximately 8% of patients with WAIHA have SLE.22 were taken into account. Other studies either found
Conversely, about 10% of patients with SLE have no association of AIHA with adverse outcomes or
AIHA.22,362-369 Budman and Steinberg370 reported that suggested that it may be associated with a more
the DAT was positive in 18% to 65% of patients in benign course.22,364,366
various published series of patients with SLE, but only Antiphospholipid Syndrome. AIHA can also be
about 10% of these patients had clinically significant part of the antiphospholipid syndrome (APS), which
hemolysis. When the DAT was positive in a patient is characterized by arterial and venous thrombosis,
whose SLE was quiescent, it was positive with anti-C recurrent pregnancy loss, and thrombocytopenia
sera but negative with anti-IgG sera; in patients with in the presence of antiphospholipid antibodies
overt hemolysis, the DAT was more commonly positive (aPL).377,378 These antibodies are identified as lupus
with both anti-C and anti-IgG sera. Rarely, the DAT was anticoagulant (LAC), which prolongs phospholipid-
positive only with anti-IgG serum. Mongan and dependent coagulation tests, or as aCL detected by
coworkers371 reported similar findings. immunoassays. This entity, first described by Hughes
AIHA may be the sole presenting sign of SLE and in 1983 in patients with SLE,379 may also appear in
predate the appearance of other disease manifesta- patients with no underlying disease, and such
tions by several years.370,372-374 Videbaek,374 for patients are diagnosed as having primary APS.
instance, cited patients in whom AIHA was present There are well-documented associations between
for 2, 3, and 6 years, respectively, before SLE was aPL and abnormalities of specific cellular components
diagnosed. More commonly, although hemolytic of the blood, such as thrombocytopenia, hemolytic
anemia is the dominant feature from the onset, symp- anemia, and leukopenia.378 Win and coworkers380
toms suggestive of SLE are also present from the start tested 474,545 normal blood donors and identified 42
of the patient’s illness. In other cases, at least some of with a positive DAT and observed 3 with positive
the signs and symptoms of SLE are established before DATs and false-positive VDRL tests, all of whom
the onset of clear-cut hemolytic anemia.243 Pirofsky2 expressed raised levels of aPL. Moreover, some
reported a simultaneous onset of SLE and AIHA in studies have demonstrated a significant correlation
five of his patients, preexisting SLE in six, and AIHA between aCL or LAC and DAT-positive hemolytic
antedating the onset of SLE in four patients, with the anemia in patients with SLE.381-386 The hemolytic
mean interval being 34 months. In some cases anemia anemia of SLE was found to be associated with IgM
may be minimal and the symptoms of SLE dominate aCL, IgG aCL, or aCL of both isotypes.381,383,384
the clinical picture; in other cases the reverse is true.243 However, the role of the aCL in the pathogenesis of
Kokori and coworkers22 reported 50 episodes of AIHA is not precisely understood.381,387
AIHA in 41 patients with SLE. They found that AIHA Kokori et al.22 found that patients with AIHA, com-
occurred as the initial disease manifestation in 27 of the pared with SLE patients with other causes of anemia,
patients (66%). A second hemolytic episode occurred in were more likely to have elevated titers of IgG aCL
seven patients (of whom six had presented with AIHA), (odds ratio [OR] = 5.8; 95% CI, 1.4 to 24) and throm-
and a third episode occurred in two patients (of whom bosis (OR = 4.6; 95% CI, 1.0 to 21). AIHA at the onset
one had presented with AIHA). The time to recurrence of SLE was independently associated with renal
was not affected significantly by any of the measured involvement (OR = 5.4; 95% CI, 1.0 to 28), thrombocy-
immunologic or clinical characteristics of SLE. topenia (OR = 7.3; 95% CI, 1.1 to 48), and possibly
Typically, the RBC autoantibodies have been thrombotic episodes during follow-up (OR = 11; 95%
reported to be warm ones. The warm antibodies CI, 0.8 to 160) compared with controls who had other
appear to differ significantly in their serologic reac- types of anemia at the onset of SLE.
tions from those found in most cases of AIHA of Fong and coworkers388 determined aCL levels in 17
idiopathic origin in that they characteristically fix Asian female SLE patients with AIHA and 29 Asian
complement and there has been no evident female SLE patients without AIHA. Both IgG and IgM
specificity.243 Although most cases of AIHA in SLE are isotypes were measured by ELISA. Elevated IgM
TABLE 3-20. SEROLOGIC DATA IN 15 CASES OF AUTOIMMUNE HEMOLYTIC ANEMIA ASSOCIATED WITH SYSTEMIC LUPUS ERYTHEMATOSUS

Antiglobulin Test Agglutination Lysis

Papainized Papainized Papainized Papainized


Direct Normal RBCs Normal RBCs
Normal RBCs PNH PNH
(titer)
Age (yr)/ Anti– Indirect (titer) RBCs RBCs Stored Fresh
Patient Sex Anti-γ G non-γ† 37°C 2–4°C 20°C 37°C 37°C 37°C 37°C 37°C Remarks

1 29/F ++ † – 16 4 4 – – – – Complement* 21 units


2 17/F +++ † ++ 16 4 16 – ++ + + Complement 15 units;
anticoagulant present
3 10/F ++ † + 128 4 4 – + – – Complement 7 units
4 24/F ++ † – 16 1 4 +++ –
5 23/F ++ † – 16 <1 4 – + + + Complement 104 units
6 26/F +++ + + 8 1 64 ++ – – Complement 16 units
7 13/M ++ + – 1 <1 1 – +++ + ± Complement 57 units
8 14/F +++ ++ – 1 <1 4 Anticoagulant present
9 15/F ++++ ++++ ++ 16 1 4 + +++ +++ + Complement 45 units
10 52/M – ± – <4 <1 1 – +± +± –
11 25/F +++ +++ ++ 8 <1 8 ++ +++ ++ ±
12 40/F ++ +± – ... <1 4 – ± – –
13 19/F +++ +++ ++ 256 4 64 ++
14 24/F +++ +++ – 16 <1 8 ++±
15 22/F ++± ++± ++++ 4 1 16 ++

* Normal range of serum complement activity = 70–150 units.


† Specific anti–non-γ serum was not used, but the reaction was only partially inhibited by the addition of human γ-globulin to the antiglobulin serum.
Modified from Dacie JV: The Haemolytic Anaemias, 3rd ed. vol. 3, Secondary or Symptomatic Haemolytic Anaemias. New York: Churchill Livingstone, 1995.

95
96 Immune Hemolytic Anemias

titers were seen in 11 (64.7%) AIHA patients and 6 Lang and coworkers392 studied aCL (IgG and IgM)
(20.7%) control patients (P < 0.01). There was no by ELISA in 74 SLE patients without AIHA, 22 SLE
significant difference in IgG aCL titers between the patients with AIHA, 50 patients with idiopathic
two groups. WAIHA, 52 patients with cold antibody AIHA, and 50
Arvieux and coworkers 389 studied sera from healthy controls. They found that the mean IgG and
patients with SLE to determine the relationship of IgM aCL titers in SLE patients without AIHA were
their antiphospholipid activity to their anti-RBC and significantly elevated compared with the values in
complement activation properties. Forty-nine per- healthy controls, and the mean aCL levels in SLE
cent of the sera had aPL as demonstrated by ELISA. patients with AIHA were higher than in SLE patients
A cellular radioimmunoassay was used for the detec- without AIHA. Interestingly, the mean aCL levels of
tion of immunoglobulin and C3 binding to erythro- patients with both warm and cold idiopathic AIHA
cytes. The authors found that the levels of IgG aPL were also significantly elevated compared with
correlated with hypocomplementemia and with healthy controls. On the basis of these data, the
immunoglobulin binding, and they concluded that authors speculated that aCL are involved in the
some aPL subsets may bind to RBCs, thus accounting pathogenesis of autoantibody-induced RBC destruc-
for the observed association of these antibodies with tion per se irrespective on an underlying SLE.
positive DATs. Latagliata and coworkers393 reported a case of APS
Hazeltine and coworkers382 detected aCL in 14 of 65 that presented with the clinical and laboratory signs of
patients (22%) with SLE using an ELISA method. Both AIHA in the absence of manifestations typically
aCL and LAC activity were associated with positive related to APS. The diagnosis of APS was made only
DATs and low serum C4 levels. Analysis of RBC after the occurrence of a sudden severe heart failure
eluates and absorption studies suggested that some due to an intraventricular thrombus requiring a surgi-
aCL may act as anti-RBC antibodies, likely directed at cal approach. The authors suggested that an accurate
membrane phospholipid epitopes. thrombophilic screening is warranted in patients with
Davidyuk and coworkers390 analyzed the plasma apparently idiopathic AIHA.
from patients with APS for their effect on the growth On the basis of these studies, one may suggest that
of allogeneic hematopoietic precursor cells using a aPL antibodies may identify a subgroup of AIHA
microtiter colony assay. They found that the IgG frac- patients different from those characterized by antibod-
tions from four patients’ plasma demonstrated ies directed against Rh-related polypeptides, band-3
enhanced BFU-E growth inhibitory activity and con- glycoprotein, and other membrane proteins.395-397
cluded that IgG autoantibodies reacting with imma- The therapy of AIHA associated with aPL antibod-
ture RBC elements may be present in patients with ies is similar to that of idiopathic AIHA. Font and
APS but that further study was necessary to deter- coworkers378 emphasized the usefulness of splenec-
mine their role, if any, in the anemia frequently seen in tomy in the treatment of refractory cytopenias (AIHA
this disorder. and/or thrombocytopenia) associated with aPL, with
Antiphospholipid Antibodies in Idiopathic a high rate of response after follow-up and no postop-
AIHA. In addition to patients who have AIHA in erative complications in reported cases.378,398
association with SLE, isolated cases have been
reported of apparently idiopathic AIHA associated
with aPL. Cabral and coworkers385 reported the pres- COLLAGEN DISORDERS OTHER THAN SLE
ence of IgM aPL antibodies in serum and on the RBCs
A number of cases describing the association of AIHA
of a patient with idiopathic AIHA during the time of
and rheumatoid arthritis have been reported.232,399-404
hemolysis but not when hemolysis abated. They
However, Estrata and coworkers232 reported that the
demonstrated that purified IgM antibody could bind
incidence of AIHA in patients with rheumatoid arthri-
to bromelain-treated RBCs and induce complement-
tis has not been shown to exceed that in the general
mediated lysis, suggesting a pathogenic role for the
population. Also, the prevalence of rheumatoid arthri-
antibody. Subsequently, similar cases have been
tis in patients with AIHA approximates that in the
reported by other investigators.389,391-393
general population. On the basis of these data, it is
Guzman and coworkers394 studied by ELISA the
difficult to establish a relationship between AIHA and
presence of aPL in sera from 18 patients with idio-
rheumatoid arthritis.232 Similarly, there are occasional
pathic AIHA and 14 patients with nonautoimmune
case reports of AIHA in association with sclero-
hemolysis. They found IgM anticardiolipin antibodies
derma.404-414
in four AIHA patients and none of the patients with
nonautoimmune hemolysis. Also, IgG aCL were
detected in six patients with AIHA and in one patient THYMOMA
with nonautoimmune hemolysis. The authors sug-
gested that the aPL do not represent a secondary phe- Thymoma, occasionally in association with pure red
nomenon caused by the release of membrane cell aplasia, has been reported to occur in patients
components but rather that they may play a role in the with AIHA, and thymectomy has been reported to be
autoimmune hemolytic process. beneficial in some such patients (see Chapter 11).
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 97

AIHA AND CARCINOMA AIHA AFTER VACCINATION


Although there are a number of reports of both Active immunization stimulates the immune system to
AIHA and carcinoma in the same subject,415-429 the produce antigen-specific humoral and cellular immu-
association is uncommon, and the question arises as nity.435 Because autoimmune diseases also involve the
to whether it is a chance event. Sokol and cowork- stimulation of the immune system against certain anti-
ers213 analyzed the incidence of RBC autoantibodies gens in the individual, it is not surprising that some con-
(with or without active hemolysis) and carcinoma in cerns have arisen as to whether immunizations may
a region with a general population of 4,662,500, and lead to the development of autoimmune diseases. A
concluded that the association was highly significant temporal association between an influenza vaccine
(P < 0.0005). Patients with RBC autoantibodies and program in the United States in 1976 and an epidemic of
confirmed carcinoma were found together 12 to Guillain-Barré syndrome caused interest to be focused
13 times more often than expected from their relative on the vaccine-autoimmunity cause-effect relationship.
frequencies. Autoantibodies occurred with a wide The incidence of Guillain-Barré syndrome in the vacci-
variety of carcinomas, with the relative incidence nated population was 5 to 6 times higher than in the
largely reflecting tumor incidence (Table 3-21). nonvaccinated population.436 More recent examples of
Further evidence for an association derives from such concerns range from whether hepatitis B immu-
reports of remission of the AIHA with removal of the nization causes multiple sclerosis to whether early
tumor,2,252,419,423-425,427,428,430-432 and its relapse with infant immunization may cause type 1 diabetes melli-
the appearance of metastases425,431,433; on specific tus.435 Evidence for immunizations leading to autoim-
absorption of autoantibody by tumor cells430; on a mune diseases come from several sources including
close temporal relation of onset of both diseases434; animal studies, single and multiple case reports, and
and on unusual age at presentation.434 epidemiologic studies. On the other hand, Offit and
Sokol and coworkers213 reviewed findings in 160 Hackett436a have indicated that well-controlled epi-
patients with carcinoma and autoantibodies, 86 of demiologic studies do not support the hypothesis that
whom had hemolysis (Table 3-22). All serologic types vaccines cause chronic autoimmune disease.
of autoantibody were found, and the ratio of The only hematologic disease for which there are
warm:cold/mixed types (7:3:1) was similar to that substantial data suggesting that immunization may
generally found in large series of patients with AIHA. be causative in some cases is immune thrombocytope-
They found that 37 (43%) had metastatic disease and nia.435,436 Nevertheless, the medical literature is filled
28 died within a few months of presentation. These with many case reports of a wide range of autoim-
findings suggest that RBC autoantibodies occur when mune illnesses temporally associated with vaccina-
there is a large tumor mass and metastatic disease, tions,435 including cases of vaccination-associated
and therefore portend a poor prognosis. immune hemolytic anemia.

TABLE 3-21. DETAILS OF PATIENTS WITH ERYTHROCYTE AUTOANTIBODIES AND CARCINOMA

No. of Age Range


Site of Primary Tumor Patients M/F (yr) Median

Breast 20 0/20 44–85 67


Lung 18 15/3 43–83 68
Pharynx 1 1/0 45
Esophagus 8 4/4 50–73 62
Stomach 12 9/3 66–89 72
Cecum 2 0/2 72–83
Colon 15 6/9 64–92 76
Rectum 15 9/6 60–85 73
Liver 1 0/1 68
Pancreas 7 2/5 61–81 64
Adrenal 1 1/0 44
Kidney 3 2/1 37–79 69
Ureter 1 1/0 70
Bladder 8 6/2 51–81 68
Prostate 14 14/0 56–83 71
Ovary 10 0/10 55–77 65
Uterus 3 0/3 59–67 65
Cervix 8 0/8 25–89 49
Skin and mucous membranes (squamous cell carcinoma) 5 2/3 62–73 69
Breast and colon (different histology) 1 0/1 51
Unknown 7 4/3 40–71 64

Modified from Sokol RJ, Booker DJ, Stamps R: Erythrocyte autoantibodies, autoimmune haemolysis, and carcinoma. J Clin Pathol
1994;47:340–343.
98 Immune Hemolytic Anemias

TABLE 3-22. SEROLOGIC PATTERN OF ERYTHROCYTE AUTOANTIBODIES IN


PATIENTS WITH CARCINOMA AND NUMBERS WITH EVIDENCE OF HEMOLYSIS

Type of Erythrocyte Autoantibodies


and No. of Patients (No. with Definite
or Strong Evidence of Autoimmune
Hemolysis)
Size of Primary Tumor Warm Cold Mixed

Breast 12 (11) 6 (4) 2 (2)


Lung 7 (6) 7 (3) 4 (4)
Pharynx 1 (0) 0 (0) 0 (0)
Esophagus 7 (1) 1 (0) 0 (0)
Stomach 9 (4) 2 (2*) 1 (1)
Cecum 1 (0) 0 (0) 1 (1)
Colon 9 (3) 5 (2) 1 (1)
Rectum 8 (7) 5 (5) 2 (2)
Liver 1 (0) 0 (0) 0 (0)
Pancreas 4 (1) 2 (2) 1 (1)
Adrenal 0 (0) 1 (1) 0 (0)
Kidney 2 (0) 0 (0) 1 (1)
Ureter 0 (0) 1 (0) 0 (0)
Bladder 6 (4) 2 (0) 0 (0)
Prostate 10 (5) 3 (1) 1 (1)
Ovary 8 (4) 2 (0) 0 (0)
Uterus 2 (0) 1 (0) 0 (0)
Cervix 5 (0) 3 (0) 0 (0)
Skin and mucous membranes (squamous cell carcinoma) 3 (1) 1 (0) 1 (1)
Breast and colon 1 (0) 0 (0) 0 (0)
Unknown 3 (2) 4 (2) 0 (0)
Total 99 (49) 46 (22) 15 (15)

* In one patient, the autoantibody was of Donath-Landsteiner type and showed evidence of specificity within the
P blood group system.
Modified from Sokol RJ, Booker DJ, Stamps R: Erythrocyte autoantibodies, autoimmune haemolysis, and carcinoma.
J Clin Pathol 1994;47:340–343.

Warm Antibody AIHA after Vaccination. A number Olcay and coworkers67 reported a 4-month-old girl
of cases of warm antibody AIHA in association with who had received BCG vaccination when 2 months
vaccination have been reported.2,13,67,104,437-444 Among old, and DPT and oral polio vaccines at 2 and again
the earliest reported cases are those of Kobl,437 who at 3 months of age. AIHA developed 1 month later, at
reported a case of AIHA after smallpox vaccination; which time the patient was admitted to the hospital
the case reported by Dameshek and Schwartz,438 with a hemoglobin of 2.7 g/dL, a hematocrit of 8.5%,
which followed poliomyelitis vaccination; and that of and a reticulocyte count of 0.2%. She received
Pirofsky,2 which occurred after vaccination with a live multiple transfusions and did not respond to
measles preparation. methylprednisolone therapy of 7 days’ duration but
More recently, Downes and coworkers443 reported a was successfully treated with a combination of
patient who developed fatal AIHA after DPT vaccina- immunosuppressive therapy consisting of cyclopho-
tion and summarized the reports of seven previously sphamide, 6-mercaptopruine, intravenous immuno-
reported cases. Including the authors’ patient, these globulin, and prednisolone. The authors did not find
cases included 3 girls and 5 boys ranging in age from an increase in the patient’s reticulocyte count
6 weeks to 7 years. Hemolytic anemia occurred in two (maximum, 0.8%) during multiple determinations
patients after the first vaccine, in four patients after during the subsequent 19 weeks even though the
the second vaccine, and in one patient after the third hemoglobin improved to between 12 and 13.4 g/dL.
vaccine. Onset of hemolysis after vaccination ranged Seltsam and coworkers444 reported a 20-month-old
from 5 days to 6 weeks. Admission hemoglobin levels girl who developed hemolytic anemia 2 weeks after
ranged from 2.3 to 9.4 g/dL (median, 4.90 g/dL). Time her third inoculation with oral polio vaccine. She
to recovery ranged from 3 weeks to 4 months. The recovered within 2 weeks, and 4 months later received
authors’ patient was admitted to the hospital in DPT vaccine without incident. However, at age 27
extremis with a hemoglobin of 2.3 g/dL and dissemi- months she developed exanthema and fever shortly
nated intravascular coagulation and was the only after a combined vaccination against mumps, rubella,
patient who had a fatal outcome. and measles. Two weeks later, she was admitted to the
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 99

hospital because of jaundice, hemoglobinuria, and Donath-Landsteiner tests became negative within 1
signs of hemolysis with a hemoglobin of 5.7 g/dL. week and she recovered promptly and completely.
The DAT was strongly positive for C3d at the time of Comments. Immune hemolysis after vaccination is
the hemolytic episodes but not between the two very rare, and serologic evidence suggesting a rela-
episodes. However, no RBC antibodies were detected tionship of the AIHA to the vaccine has been reported
at any time. She was treated with antibiotics, pred- only once. Haneberg and coworkers441 reported anti-
nisolone, and blood transfusions and recovered bodies to tetanus and diphtheria toxoids and pertus-
within 2 weeks. sis in heat eluates from one infant suggesting that
A second patient reported by Seltsam and cowork- these antibodies were bound to the RBCs. However,
ers444 was a 21-month-old boy with an uneventful available data are certainly insufficient to indicate
medical history who was inoculated with a combina- either the presence or absence of a causal relationship
tion of six vaccines, including the fourth doses of DPT between vaccination and hemolytic anemia.446
and Hemophilus influenzae vaccines, and the third Although the reported cases could all be coinciden-
against polio and hepatitis B. Four days later, he tal because vaccination is such a common event, a
became pale and jaundiced and was admitted to the cause-and-effect relationship cannot be ruled out.
hospital with a hemoglobin of 3.7 g/dL. He was
treated with prednisolone and recovered. The DAT AIHA AND INFECTIOUS AGENTS
was strongly positive using anti-IgG, and weakly pos-
itive using anti-C3d. The authors give few serologic Although AIHA in association with upper respiratory
details stating only that the IAT was positive with infections and unspecified “viral” syndromes or “flu-
RBCs tested (11 panel cells) and at a follow-up 4 like illness” is common, the incidence of AIHA after
months later RBC antibodies were not detected in the well-documented specific infections is relatively
serum or eluate. uncommon. In the series of 1223 patients reported by
Other Types of AIHA after Vaccination. Martinez Sokol and Hewitt,208 infectious mononucleosis and
and Domingo445 reported a case of Evans’s syndrome mycoplasma pneumonia were the only two infectious
apparently triggered by recombinant hepatitis B diseases that were documented in more than three
vaccine. Two days after receiving his second dose of patients. Nevertheless, there are well-documented
hepatitis B vaccine, a 33-year-old man, who was a instances in which viral, bacterial, parasitic, and other
chronic HbsAg carrier, presented with acute abdomi- infectious agents have been associated with immune
nal pain, polyarthralgia affecting his hands and feet, hemolytic anemia. (Infections associated with PCH
chills, and fever. He had had no reaction to the first were reviewed earlier.)
dose of vaccine. His hemoglobin was 5.0 g/dL, the There are a number of mechanisms by which infec-
reticulocyte count was 20%, and the platelet count tions may result in hemolysis on an immune
was 11,000/μL. The DAT was positive using anti-IgG basis.447,448 Among these are that the infection may
and anti-C3 sera. The authors did not report the pre- result in the production of either cold- or warm-react-
sence or absence of red cell antibodies but did find cir- ing RBC autoantibodies; antibody-antigen complexes
culating immune complexes and IgG platelet-bound specifically related to the infectious agent may coat
antibodies suggesting an immune mechanism for the the RBCs, which then act as an “innocent bystander”;
thrombocytopenia. The patient was treated with corti- the infectious agent may result in exposure of RBC
costeroids and recovered over a period of 2 months. antigens, which are normally hidden, to naturally
Bunch and coworkers169 described a 19-month-old occurring antibodies (polyagglutination).
girl who, 6 weeks before her admission, was immu- Seitz and coworkers,448 using an immunofluores-
nized with live attenuated measles vaccine. Three cence test, studied 96 children with hemolytic anemia
weeks later she developed a mild illness with a mor- and found that the RBC membrane was altered in vivo
billiform rash, conjunctivitis, and rhinorrhea, and in a majority of patients by nonspecific adsorption of
during the next week she also developed hemoglo- foreign material released from infectious micro-
binuria. On admission the hemoglobin was 3.3 g/dL, organisms. In addition, additive binding of complement
reticulocytes 5%, and platelets 498,000/μL. The DAT was detectable by the antiglobulin test. Accordingly,
was strongly positive. A diagnosis of paroxysmal cold they suggested the adsorption of microbial antigens to
hemoglobinuria was made because the serum con- the RBC surface as one of the causes for RBC destruc-
tained a typical Donath-Landsteiner antibody charac- tion during infection-associated hemolytic anemia.
terized as a biphasic, complement-fixing IgG antibody Additional details regarding the mechanisms by
with anti-P specificity. The antibody sensitized RBCs which autoantibodies are produced in response to
up to 25°C. Syphilis was excluded by negative viral infection have been reviewed by Morrow and
Wassermann reactions and VDRL screening tests in coworkers449 and by Meite and coworkers.450 Experi-
the patient and both of her parents. The measles com- mental data have suggested that viruses trigger an
plement-fixing antibody titer was high at a level of autoimmune humoral response by a number of
128. The patient was kept warm and transfused with distinct mechanisms, including polyclonal B-lympho-
RBCs warmed to approximately 37°C. The DAT and cyte activation, antigenic mimicry, modification of
100 Immune Hemolytic Anemias

self-antigen, production of anti-idiotypic antibodies, failure,466-468 nephrotic syndrome,469 mucocutaneous


or enhancement of major histocompatibility complex lesions including erythema multiforme and Stevens-
molecule expression on potential antigen-presenting Johnson syndrome,470 disseminated intravascular
cells. They also presented data indicating that viruses coagulation, myocarditis, pericarditis, myositis, pol-
can trigger the onset of AIHA by enhancing the path- yarthritis, meningoencephalitis, cranial and periph-
ogenicity of autoantibodies. This observation may eral neuropathy, hepatitis, and pancreatitis.471
indicate how different viruses can trigger similar clin- In recent years postmycoplasma hemolytic anemia
ical autoimmune diseases.450 seems to have been reported less frequently.459 In part
Specificity of Autoantibodies in AIHA Secondary this may be due to the fact that the disorder is no
to Infectious Agents. In postinfection cold antibody longer the novelty it was in earlier years, but also,
AIHAs, there is a striking relation between the effective antibiotic treatment of the infection has
specificity of the cold antibody and the underlying perhaps diminished the incidence of hemolysis.
infectious agent.451 Cold antibodies of anti-I specificity Nevertheless, case reports continue to appear in the
are found in infections with M. pneumoniae, whereas medical literature.455,470,472-474
cold antibodies of anti-i specificity frequently occur in Hemolysis is self-limiting and usually resolves in
infections with EBV. AIHA associated with varicella or 2 to 3 weeks. However, the hemolytic anemia may
rubella infections have commonly been reported to proceed at an alarming rate and, indeed, fatalities
have anti-Pr specificity. However, exceptions to these have been reported.475,476
findings have also been reported, as indicated later. Although AIHA associated with M. pneumoniae
Mycoplasma pneumoniae Infections. A rise in the infection is almost always caused by cold agglultinins,
cold agglutinin titer occurs frequently after M. pneu- Louie and coworkers477 reported a patient with severe
moniae infection. From 33% to 76% of patients with hemolysis who had both a high titer, high thermal
M. pneumoniae infection have a cold agglutinin titer amplitude cold agglutinin, and an IgG warm agglu-
greater than 64 or have a fourfold or greater increase tinin. The DAT was strongly positive with anti-IgG
in titer during convalescence.452,453 An acute cold and anti-C3, and the patient’s hemolysis persisted
agglutinin response is sometimes seen in viral infec- despite declining thermal amplitude and titer of the
tions such as influenza A and infectious mononucleo- cold agglutinin. The DAT remained positive with anti-
sis, but very high titers are seen almost exclusively in IgG, and there was an excellent response to pred-
mycoplasma infections. Usually the magnitude of the nisone, leading the authors to conclude that the warm
cold agglutinin response is related to the clinical IgG antibody was responsible for the patient’s AIHA.
severity of the pneumonia,452-454 although severe Mechanism of Development of Anti-I in Myco-
anemia has been reported in association with clini- plasma pneumoniae Infection. The cold agglutinin
cally mild pneumonia.455 produced in response to human infection with
Although overt hemolytic anemia is uncommon in M. pneumoniae, as well as the antibody found in idio-
patients with M. pneumoniae infection, subclinical pathic CAS, usually have specificity for the blood
hemolysis may be rather common as indicated by the group I antigen contained in red cell glycoproteins.
fact that a significant fall in hemoglobin concentration The mechanism by which cold agglutinins are elicited
(>2 g/dL) has been observed in 12.8% of patients in by M. pneumoniae has been studied by several groups
one series,456 and elevated reticulocyte counts (>2%) of investigators. One proposed mechanism is that the
were found in 64% of infections.457 A small minority organism itself possesses an I-like antigen that stimu-
of patients respond to the infection in an exaggerated lates the formation of cross-reacting anti-I antibodies.
way, that is, they produce cold antibodies of unusu- An alternative hypothesis is that the organism acts on
ally high titer and which react at an unusually high and modifies the I antigen on the RBC membrane so
temperature.458 When hemolytic anemia occurs in that it becomes unusually autoantigenic. Evidence has
association with M. pneumoniae infection, it does so been presented to support both hypotheses.
usually in the second or third week of the patient’s Schmidt and coworkers478 found that 18 of 25 strains
illness. The patient may already be recovered from his of mycoplasma blocked or destroyed the I antigen
respiratory infection, but becomes ill once more with receptors on normal RBCs as judged by subsequent
increasing pallor and jaundice.459 A rapid onset of agglutinability of the cells by an anti-I present in the
hemolysis is frequently observed, and abnormal IgM serum of an I-negative individual. These investigators
cold agglutinins (which characteristically have anti-I also inoculated 27 volunteers with M. pneumoniae
specificity) are present. Exposure to cold may precipi- vaccine and found that the thermal amplitude of anti-I
tate hemolysis.109 The hemolysis may be severe,460 in their serum was increased in 12 and the titer of the
and even life threatening.455 Splenomegaly is gener- anti-I was increased in 4.479 Feizi and coworkers480
ally present; acrocyanosis and hemoglobinuria are found that 9 of 28 (32%) rabbits inoculated with OI cells
unusual,2 although massive hemoglobinuria with and M. pneumoniae developed fourfold or greater rises
acute intravascular hemolysis has been reported.461 in cold-agglutinin titers. Loomes and coworkers481 sug-
Gangrene of the extremities is rare but has been well gested that anti-I antibodies might arise in M. pneumo-
described in the older medical literature.462-465 Other niae infection in response to a modification of the
unusual manifestations of the disease are renal “self”-antigen-I as a result of its interaction with the
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 101

organism. They reported that the interaction of M. Clinical Findings. Acute pharyngitis often brings
pneumoniae with human erythrocytes is mediated by the patient to the physician. The classic features of the
long chain oligosaccharides of sialic acid joined by disorder are fever lasting up to 3 weeks or more, lym-
alpha 2-3 linkage to the terminal galactose residues of phadenopathy, splenomegaly, and florid, atypical
poly-N-acetyllactosamine sequences of Ii antigen type. lymphocytosis along with malaise that may be slow to
In contrast, several groups of investigators pre- resolve. Coupled with a positive heterophil antibody
sented evidence that M. pneumoniae does bear an I-like reaction or “Monospot test,” these features are
antigen. Costea and coworkers353,482 injected suspen- enough for diagnosis in most cases. The diagnosis is
sions of M. pneumoniae into rabbits and found that verified by the presence of EBV antibodies.493
they stimulated the formation of cold agglutinins. Some of the clinical features of hemolytic anemia
They also reported that the cold agglutinins thus occurring in association with infectious mononucleo-
formed reacted with the micro-organism as well as sis are listed in Table 3-23. The signs of hemolysis
with the animals’ RBCs. This group of investigators usually develop 1 to 2 weeks after the onset of the
also found that, although intact M. pneumoniae organ- illness, although in some cases features of infectious
isms were unable to absorb cold agglutinins from the mononucleosis and hemolytic anemia develop simul-
sera of patients who had mycoplasma pneumonia, a taneously. Most of the patients who develop AIHA are
mycoplasma lipopolysaccharide was able to reduce at first acutely ill, with high fever, and then become
titers sixfold or more. They concluded that the anti- weak, anemic, and jaundiced, and may have hemo-
gens in M. pneumoniae with which cold agglutinins globinuria.494,495 About 74% of patients have a palpa-
react are hidden in the organism’s limiting membrane ble spleen and an enlarged liver.496
and that the development of cold agglutinins in man Anemia is usually mild and self-limiting,497
is due to immunization by cross-reacting antigens.483 although severe, life-threatening hemolysis has been
These conclusions were supported by the later work reported.495,497,498 The peripheral blood film may
of Lind484 and Janney and coworkers.485 contain spherocytes and, in unusual instances,
Management. Management includes keeping the autoagglutination and erythrophagocytosis.499 The
patient warm. This is particularly important since WBC count is generally above normal and a varying,
chilling is likely to cause an increase in hemoly- but often large, proportion of the cells are abnormal
sis.108,109,459 In patients with severe anemia, the hospi- lymphocytes typical of infectious mononucleosis.
tal room should be warmed to as high a temperature Palanduz and coworkers500 reported a 7-year-old
as is tolerable, and mittens and warm slippers worn as girl who developed AIHA and fulminant hepatic
well. One group even resorted to the use of a NASA failure associated with EBV infection; the patient
space suit to provide constant warmth in a rare recovered after corticosteroids and supportive
patient with profound disease.486,486a If blood transfu- therapy.
sion becomes necessary, the principles and techniques Several patients have been described who devel-
reviewed for CAS in Chapter 10 should be followed. oped signs of overt hemolysis during an attack of
Corticosteroid therapy is usually not indicated infectious mononucleosis and were subsequently
because such therapy is generally ineffective in CAS found to have previously inapparent hereditary
and the hemolysis frequently is not severe and can be spherocytosis.501-506 Except in the patient reported
expected to subside spontaneously. Nevertheless, by DeNardo and Ray,503 evidence of an autoimmune
several patients with severe hemolytic anemia due to cause of the hemolysis was lacking. Instead, the
mycoplasma-induced cold agglutinin disease have hemolysis was attributed to the fact that infections,
responded to high-dose corticosteroids.471,472,487,488 perhaps of any type, are a well-known cause of exac-
Tetracycline, erythromycin, and other antibiotics erbation of hemolysis in hereditary spherocytosis.
are effective against M. pneumoniae,489 but at the time
hemolysis begins, the pneumonia may be resolved.
Nevertheless, in some patients who delay seeking
medical attention for their respiratory symptoms, TABLE 3-23. CLINICAL FINDINGS IN HEMOLYTIC
antibiotic therapy has seemed to promptly resolve ANEMIA ASSOCIATED WITH INFECTIOUS
both the pneumonia and the hemolytic anemia (see MONONUCLEOSIS
Chapter 11).
Infectious Mononucleosis (Epstein-Barr Virus). Onset of hemolysis in relation to 1 wk (28%)
Infectious mononucleosis is a common disorder, and the onset of infectious 7–13 days (44%)
AIHA developing during its course is a well-recog- mononucleosis 14–21 days (15%)
nized entity. However, AIHA does not occur com- 21 days (13%)
Duration of hemolysis <1 mo (71%)
monly, and Dacie and Worlledge5 concluded that 1–2 mo (25%)
fewer than 1:1000 patients with infectious monucleo- >2 mo (4%)
sis develop overt hemolysis. Einzig and Neerhout490 Enlargement of spleen Palpable (74%)
pointed out that acute hemolytic anemia was not Non palpable (26%)
Enlargement of liver Palpable (74%)
noted in five large series comprising 1113 patients. Non palpable (26%)
Other studies indicate an incidence of 2% to 3%.491,492
102 Immune Hemolytic Anemias

Serologic Findings. The DAT is generally positive, The Donath-Landsteiner reaction was positive, and it
although some cases with a negative DAT have been remained so for at least 7 weeks. The antibody did not
described.497,498 The sera often contain a cold antibody have any identifiable specificity. Burkhart and Hsu177
that agglutinates cord-blood RBCs more strongly than described a 23-year-old man in whom the Donath-
adult RBCs, that is, anti-i. In 1965, Jenkins and cowork- Landsteiner reaction was positive. The antibody in his
ers507 and Calvo and coworkers508 reported the first serum was identified as anti-i and was an IgM globu-
cases of hemolytic anemia in infectious mononucleosis lin. The titer at 4°C with i (cord) RBCs was 16,384 and
apparently mediated by the temporary production of a with I RBCs was 512. At 22°C the titer with the i RBCs
high thermal amplitude cold agglutinin of anti-i was 64; at 32°C it was zero. The cold-warm (4°C to 37°C)
specificity. The latter authors also described the pres- procedure using i cells was positive to a titer of 64.
ence of anti-i cold agglutinins in 23 of 38 uncomplicated Management. Management of patients with AIHA
cases of infectious mononucleosis. Other reports associated with infectious mononucleosis is reviewed
confirmed and extended these findings.496,509-513 in Chapter 11. Of note here is the variable response to
However, the finding of anti-i of high thermal ampli- blood transfusion that has been reported. The patients
tude is not a uniform finding in patients with AIHA of Silber and coworkers497 and Tonkin and cowork-
associated with infectious mononucleosis. Wilkinson ers498 had severe hemolysis, but no problems were
and coworkers514 reported three patients and empha- encountered with blood transfusion. In contrast,
sized how variable the antibody pattern might be. The Perkin and coworkers517 reported that their patient
serologic findings in the first patient were considered to received five units of compatible blood and had
be characteristic: his serum agglutinated cord i RBCs to symptoms of a hemolytic transfusion reaction, includ-
a titer of 512 at 4°C and to titers of 64 at 22°C and 4 at ing back pain, dyspnea, and fever. Furthermore, his
31°C; it also agglutinated adult I RBCs to a titer of 64 at hemoglobin concentration increased by only 3.4 g/dL.
4°C and 2 at 22°C. The two other patients had formed Cytomegalovirus Infection. Zuelzer and associ-
anti-i antibodies, but they were of low titer at 4°C and ates518,519 were among the first to emphasize the rela-
did not cause agglutination at 22°C. The fact that the tionship between cytomegalovirus (CMV) infection
antibodies were not reactive at physiologic tempera- and hemolytic anemia. They studied 22 children with
tures made it unlikely that they were the cause of the CMV, 18 of whom had positive DATs, and concluded
patients’ AIHA. In none of the patients were warm that the presence of CMV in immunohemolytic
autoantibodies detected. anemia was not fortuitous. They described an associ-
Woodruff and McPherson495 described a patient ation between episodes of lymphadenitis due to CMV
who had developed severe intravascular hemolysis. and periods of hemolysis. However, not all patients
His serum agglutinated both i cord and I adult cells to developed autoantibodies, and they suggested that
titers at 4°C of 128 and 64, respectively; at room RBC antibody production was a secondary phenome-
temperature, the titers were 16 and 32, and at 37°C, non without clinical significance.
0 and 4. Subsequent reports of CMV infection with associ-
Lee and coworkers515 suggested that the Lewis ated hemolytic anemia have also been variable
status of the patient is important in the development regarding the presence or absence of RBC autoanti-
of hemolysis in infectious mononucleosis. They made bodies. Kantor and coworkers520 recorded the clinical
this suggestion because of finding two patients with and immunologic findings in 10 patients who had
infectious mononucleosis and AIHA who were Le(a− developed postperfusion CMV infection; three of
b−), and two other patients with a history of infectious them developed transient hemolytic anemia. The DAT
mononucleosis and AIHA who were Le(a−b−) and was positive in two of seven patients tested, and the
Le(a+b−), respectively. Of the four patients, three were cold agglutinin titer was raised to 512 and 2048 in two
nonsecretors and the fourth likely to be so. The patients. Harris and coworkers521 and van Spronsen
authors pointed out that the Lewis status of these four and Breed522 described patients with CMV-associated
patients varies significantly from that of other patients hemolytic anemia and thrombocytopenia, but DATs
with uncomplicated infectious mononucleosis and were negative and there were no RBC antibodies in
from the known distribution of the Lewis type in the the patients’ sera.
white population. Although hemolytic anemia with a positive DAT in
Bowman and coworkers499 reported the presence of association with spontaneous CMV infection is rare in
a potent cold-reacting auto-anti-N with increased adults, Salloum and Lundberg523 reported two such
thermal amplitude in the serum of a patient with patients and reviewed the findings in six patients
severe immunohemolytic anemia after infectious reported by others.524-527 Although the DAT was pos-
mononucleosis. itive in all patients, serum antibodies were found in
Patients in whom the Donath-Landsteiner reaction only two of the eight cases.524,527 The authors sug-
has been positive have been reported by several gested that the true incidence of immunohemolytic
groups of investigators.176,177,516 Wishart and anemia associated with CMV infection may be under-
Davey176 reported a 17-year-old male whose serum estimated because CMV serology is not routinely
contained a high-thermal-amplitude cold antibody obtained as a part of hemolysis work-up. Subsequently,
which agglutinated RBCs to a titer of 256 in the cold. Gavazzi and coworkers528 reported an unusual case of
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 103

primary CMV infection manifested by severe hemoly- infection. The DAT was positive using anti-IgM, anti-
sis in an immunocompetent adult. The responsible C, and anti-IgA, but negative using anti-IgG. The
mechanism for the hemolysis was unclear because no serum contained anti-i to a titer of 128 at 4°C, 32 at
RBC antibodies were demonstrated and the DAT was 22°C, and 1 at 37°C. The anti-i disappeared when the
positive “with a low titer” due to complement and complement-fixation antibody titers against CMV
IgG. The authors emphasized that severe, multiorgan decreased. Berlin and coworkers531 reported a patient
CMV infection may occur even in immunocompetent with spontaneous CMV infection who developed
adults,529 but hemolysis seems to be rare even in these hemolysis and whose serum reacted with cord ery-
patients. throcytes to a titer of 128 and reacted more weakly
Horwitz and coworkers527 reported two patients against adult RBCs. However, the DAT was negative
with hemolysis in previously healthy adults with and no thermal amplitude studies were performed so
CMV infections. In one patient, the DAT was positive that the significance of the serum antibody is obscure.
using anti-IgG serum, but the only serum antibody CMV is also only rarely found in children with
was a weakly reactive low-molecular-weight cold AIHA. CMV infection was not found among the
agglutinin. In the other patient, the DAT was negative 42 children with AIHA reported by Sokol and cowork-
and no RBC autoantibodies were found. The authors ers,11 and Habibi and coworkers13 found CMV infec-
reviewed data from 20 additional patients with CMV tion in only 1 of 80 children. The patient reported by
infection who had been studied in their laboratory McCarthy and coworkers534 was complicated by
during an 11-year period. They concluded that there CMV infection, although how significant the infection
had been evidence for subclinical hemolysis in at least was in causation hemolysis is unclear. Murray and
50% of patients. DATs had been carried out on 10 coworkers535 described two infants with severe CMV-
patients and were positive in only 3. Ten patients had associated WAIHA. The authors demonstrated anti-
cold-agglutinin titers of less than 56, but in no case CMV IgG antibody in one case and suggested a
was the thermal amplitude reported. The authors con- possible causal relationship between AIHA and CMV
cluded that the mechanism responsible for hemolysis infection. Both patients were ultimately treated with
was obscure. intravenous CMV immune globulin, with subsequent
Dietz530 reported a 21-year-old man who developed improvement. Hausler and coworkers536 reported a 9-
fever, fatigue, precordial chest pain, marked shortness month-old infant with congenital HIV-1 infection who
of breath, and dyspnea on minimal exertion. He had presented with severe AIHA and a high CMV viral
enlarged lymph nodes in the inguinal and axillary plasma load. After successful therapy of hemolysis,
areas, an enlarged liver, and a palpable spleen. CMV CMV was not detected in the plasma, which sug-
titers (determined by complement fixation) showed gested a causal relationship between the hemolysis
an increase from 8 to 512, and the titer of CMV-IgM and CMV infection.
antibodies was 128. Cultures of a buccal smear and of Human Immunodeficiency Virus. Anemia occurs
urine on human fibrobast monolayer medium grew frequently among patients seropositive for HIV, but its
CMV. He developed hemolytic anemia with a drop in multifactorial origin complicates its differential diagno-
hematocrit from 48.5% to 35% with a reticulocyte sis.537-539 The most frequent form of anemia in AIDS
count of 8.5%. During the episode of hemolysis, the has the characteristics of anemia of chronic
DAT was positive, but no results of tests for serum disease.538,540 Other causes include opportunistic infec-
antibodies are described. Similarly, Berlin and tions, B19 parvovirus infection, myelosuppressive
coworkers531 reported an adult woman who devel- drugs, thrombotic thrombocytopenic purpura, hyper-
oped CMV and hemolytic anemia with the hemoglo- splenism, marrow infiltration with tumor, iron
bin falling from 12.3 to 8.3 g/dL. However, serologic deficiency, and blood loss anemia.538,540,541 Also,
results are incompletely documented; the DAT was endogenous erythropoietin concentrations are fre-
negative, and the IAT was negative against adult quently low and reticulocytopenia is a common
RBCs but “weakly reactive” against human cord finding.538 In addition, many autoimmune aberrations
RBCs. On this basis the authors assumed that the have been described in individuals with AIDS.449
“anti-i” initiated the hemolytic process. Although some of these abnormalities, such as hyper-
Reller532 reported a patient with hemolytic anemia gammaglobulinemia and the presence of circulating
that developed during a CMV infection in a previ- immune complexes, are only generalized indicators of
ously healthy 30-year-old woman. There was an faulty immunoregulation or even a direct consequence
abrupt fall in hematocrit followed by an increased of infection, more specific clinical and pathologic evi-
reticulocyte count and erythroid hyperplasia in the dence of autoimmunity, including the development of
bone marrow. The patient’s cold agglutinin titer AIHA, has been associated with HIV infection.
increased from 16 to “at least” 512 during the illness Prevalence of Positive Direct Antiglobulin Tests.
and demonstrated anti-I specificity. Results of Most reports indicate that patients with AIDS com-
antiglobulin tests were not provided. monly have a positive DAT.542-546 The positive DATs
Aguado and coworkers533 reported a patient with may be due to hyperglobulinemia,542 circulating
multiple myeloma and monoclonal cryoglobulinemia immune complexes,544,547 or anti-RBC autoantibod-
who developed severe hemolytic anemia after CMV ies.537,543,548 Toy and coworkers542 reported that the
104 Immune Hemolytic Anemias

prevalence of a positive DAT was 18% (10 of 55) in Some reported cases of immune hemolysis in asso-
AIDS patients compared with 0.6% in general hospi- ciation with HIV infection have been caused by drug
tal patients during a 2-year period. In the series of administration. Fatal acute immune hemolysis has
Zon and coworkers,544 the prevalence of a positive been caused by ceftriaxone565 and has also been
DAT in patients with HIV antibodies was 21%, and reported in a patient receiving indinavir, a protease
McGinnis and coworkers543 found positive results in inhibitor.566 In the latter case, the causation of immune
12 of 28 (43%) patients with AIDS. hemolysis by the drug is not clear because hemolysis
Bordin and coworkers545 tested blood samples on did not appear until 10 months after therapy was
239 patients with HIV infection and found a positive begun, and no serum red cell antibodies were
DAT in 16.7%. They further studied 67 patients using described. The drug was implicated nonetheless
an enzyme-linked antiglobulin test to measure more because of a positive DAT caused by complement, and
accurately the number of IgG molecules per RBC. because of relapse of hemolysis within 24 hours after
They found that 30 of the 67 individuals had increased reintroduction of the drug. The authors suggested
numbers (mean, 155) compared with normal controls caution in the use of protease inhibitors in such
and with patients with hypergammaglobulinemia patients (see Chapter 8).
due to multiple myeloma or chronic liver disease. The Diagnosis and Management. The diagnosis of AIHA
authors suggested that some AIDS patients may have may be missed in patients with AIDS because anemia
specific binding of IgG on the surface of their RBCs, of other causes is very common, whereas AIHA is
rather than nonspecific uptake. uncommon.539 Also, reticulocytopenia may tend to
In contrast, Lepennec and coworkers549 found posi- obscure the diagnosis. A high transfusion requirement
tive DATs in only 14 of 185 (7.5%) individuals with may be an important indication of the development of
asymptomatic HIV infection, and van der Lelie and AIHA (see Chapter 2), and RBC autoantibodies may
coworkers550 detected no RBC-bound immunoglobu- first be discovered by the transfusion service.
lins in any of their 16 patients, although platelet- Treatment of AIHA in HIV-infected patients has
bound immunoglobulins were demonstrated by generally been successful.537 Patients may respond
immunofluorescence in all of the patients, and granu- well to corticosteroid therapy,220,537,548,555,557 splenec-
locyte-bound immunoglobulins were found in 12. tomy,537 zidovudine,559 and blood transfusion.556,557
Immune Hemolytic Anemia. Despite the frequent Reports of an increased incidence of thromboem-
occurrence of a positive DAT, AIHA is uncommon bolic events in HIV-positive persons567-571 suggest
in patients with HIV infection.220,548,549,551,552 that there may be an additional hazard of transfusion
Nevertheless, a number of case reports have been therapy in these individuals. Hemolysis results in
published.32,33,536,537,548,553-564 Most patients have had generation of thromboplastic substances from the
WAIHA, although some reports included patients erythrocyte stroma and may lead to intravascular
who had cold antibodies537,555,557,560-562 or both cold coagulation.572 (Also see pages 64–66 for a discussion
and warm antibodies.220 of thrombotic events in AIHA.) The increased volume
Immune hemolysis may be clinically and serologi- of circulating RBCs after transfusion of a patient with
cally indistinguishable from idiopathic WAIHA,548 AIHA will result in an increased amount of RBC
although some special features deserve comment. destruction even though the rate of cell destruction is
Reticulocytosis, which is characteristic of non-AIDS– unchanged. In patients with a hypercoagulable state,
related AIHA, is often lacking in HIV-infected this may lead to thrombotic events. Indeed, Saif and
patients537,538,540,556,560,564 despite bone marrow ery- coworkers32 reported a patient with HIV-associated
throid hyperplasia.537,564 The levels of erythropoietin AIHA who developed a pulmonary embolism after
may be low and therapy with erythropoietin may be transfusion of one unit of RBCs. Although the pul-
of significant benefit.538,540 monary embolism may well have been a coincidental
Koduri and coworkers221 reported four men with finding, the authors cited two other reports of throm-
advanced HIV infection (AIDS) and AIHA (Table 3-24). boembolism in patients with HIV-associated
All patients presented with an acute onset of severe AIHA.26,573 They also cited the case of Bilgrami and
AIHA, fever, and splenomegaly. The DAT was positive coworkers,33 although the latter report described DIC
in all four patients using anti-IgG, and was positive in rather than thromboembolism.
three patients with anti-C3. The IAT was positive in all, Varicella (Chickenpox). AIHA is a very rare com-
and the serum and red cell eluate showed a pan- plication of chickenpox, although a number of such
agglutinin in each patient. Three of the patients were cases have been reported.574-581
reported to have had mixed warm and cold autoanti- In most instances of varicella-associated AIHA,
body hemolytic anemia, based on the fact that there the hemolysis develops after the onset of chicken-
was strong agglutination of reagent RBCs at 22°C, pox.575-579 Northoff and coworkers579 described the
although no thermal amplitude studies were per- occurrence of a transient attack of acute hemolysis
formed. Shortly after admission, two patients died of affecting a 43-year-old woman during the acute phase of
severe anemia. Two patients responded to prednisone disease. Her hemoglobin fell from 14.4 to 11.1 g/dL, and
therapy and were in remission from AIHA for 15 and her serum contained a monotypic IgG-κ cold antibody
30 months, respectively, at the time of the report. which had anti-Pr1h specificity. The DAT was positive
TABLE 3-24. CLINICAL AND LABORATORY DATA IN FOUR PATIENTS WITH AIDS AND AUTOIMMUNE HEMOLYTIC ANEMIA

Temp CD4 DAT CA Hb MCV Reticulocytes White Blood Platelets TB/DB LDH
Patient Medications* (°C) (×106/L) IgG C3 (titer) (g/dL) (fL) (%) Cells (×109/L) (×109/L) (mg/dL) (IU/L)

1 None 39.0 10 4+ + 160 2.0 119 11 16.8 130 3.0/0.5 642


2 ZDV, 3TC 38.9 120 4+ Neg† 4+ 3.4 97 18 7.3 149 2.4/0.4 916
3 D4T, 3TC, 38.3 55 4+ + Neg 4.7 117 32 15.3 174 6.4/0.9 916
RIT, SAQ
4 ZDV, 3TC, 38.9 13 3+ 3+ 4+ 4.1 124 23 17.2 324 7.5/3.7 722
RIT, SAQ

Temp, body temperature on admission; CD4, CD4+ T-lymphocyte count; IgG, direct antiglobulin test using IgG antiserum; C3, direct antiglobulin test using C3b–C3d antiserum; CA, cold agglutinins; MCV, mean cell
volume; TB, total serum bilirubin; DB, direct serum bilirubin; LDH, serum lactate dehydrogenase; Neg, negative.
* Antiretroviral medications at the time of admission: ZDV, zidovudine; 3TC, lamivudine; D4T, stavudine; RIT, ritonavir; SAQ, saquinavir.
† C3d Coombs negative.
Modified from Koduri PR, Singa P, Nikolinakos P: Autoimmune hemolytic anemia in patients infected with human immunodeficiency virus. Am J Hematol 2002;70:174–176.

105
106 Immune Hemolytic Anemias

with anti-C but negative with anti-IgG, -IgA, and Rubella. Konig and coworkers451 reported a 5-year-
-IgM. The patient’s serum agglutinated all RBCs old boy who developed severe hemolytic anemia after
tested at 22°C, but not at 37°C; the antibody titer at serologically ascertained rubella infection. The hemo-
0°C was 256. globin dropped to 4.2 g/dL, and he required RBC
In contrast, the patient reported by Friedman and transfusion. Although the DAT was negative through-
Dracker576 developed AIHA in the convalescent phase out the disease, the authors attributed the hemolysis
of his illness, and the patient reported by Terada to the presence of an IgG-λ-monotypic cold antibody
and coworkers574 developed AIHA during the incuba- with anti-Pr specificity.
tion period. The latter patient was a 2-year-old boy Kadota and coworkers584 described a 24-year-old
who was admitted to the hospital with a hemoglobin man who presented with dark urine and general
level of 7.3 g/dL, a reticulocyte count of 8.2%, and other malaise. On the fourth day of illness general exan-
laboratory evidences of hemolysis. On the second day thema and retroauricular lymph nodes were recog-
after admission, chickenpox vesicles were found mainly nized. Although the exanthema disappeared, signs of
on his trunk, and the patient was treated with acyclovir. hemolysis requiring hospitalization became apparent.
The course of both the chickenpox and the hemolytic Rubella was confirmed by the presence of high titer
anemia were mild. The case is unusual in that the rubella antibodies, and cold agglutinins were present
specificity of the cold antibody was anti-I. The patient to a titer of 2048. However, as in the case of Konig and
reported by Johnson578 also had a benign course and coworkers, the DAT was negative.
recovered after a short course of corticosteroids. A series of 13 patients with hemolytic anemia after
The only fatal case was reported by Herron and rubella were described by Ueda and coworkers,585 but
coworkers,577 although the significance of the AIHA there is little documentation of an immune cause of
in regard to her demise is uncertain. Their patient was the hemolysis. The indirect antiglobulin test was
a 39-year-old woman who had an extensive exanthe- positive in only two of the 13 cases, and the DAT was
matous rash and varicella pneumonia with wide- positive in only 3 of the 11 cases in which it was per-
spread pulmonary infiltrates in the chest radiograph. formed.
On admission. there was no evidence of hemolysis, Miyazaki and coworkers586 reported a 7-month-old
her hemoglobin was normal, and no RBC antibodies patient with congenital rubella who had a hemoglobin
were found. Three days later her hemoglobin dropped of 6.8 g/dL and a positive DAT which revealed IgM
to 9.1 g/dL and cold agglutinins of anti-Pr specificity and C3 on the RBCs. The IAT was also said to be pos-
were found with a titer of 128 at 0°C. She ultimately itive although no details are provided. Also, the LDH
developed multiorgan failure and died 3 weeks after was 7760 units (normal range, 100 to 200 units).
admission. However, the reticulocytes were 0.1%, and the bone
In the case reported by Parashar and coworkers,580 marrow revealed a marked paucity of erythroid cells
the association of AIHA with chickenpox seems less so that the evidence for hemolysis as a significant
certain. These authors reported an Indian boy who cause of the patient’s anemia is inconclusive. The
developed persistent fever with progressive jaundice patient was treated with corticosteroids and transfu-
1 month after having chickenpox. After 1 month of sions and died at 9 months of age.
these symptoms, he was admitted with severe DAT- Cold antibodies of anti-Pr specificity have com-
positive AIHA. monly been associated with infections with rubella
Terada and coworkers574 reviewed reports of chick- infection.451,587,588 Konig and coworkers451 reviewed
enpox and hemolytic anemia and found four patients the reports of seven patients with rubella and cold
who had anti-Pr cold agglutinins.576-579 Other specifi- antibodies and found that anti-Pr specificity was
cities that have been reported include “complete anti- present in six451,587-592 (Table 3-25). An additional case
body,”575 “warm and cold antibody,”580 biphasic was reported by Kadota and coworkers.584
anti-P (Donath-Landsteiner antibody),581 anti-DC,582 Parvovirus B19. AIHA is among the least common
and anti-I.574 of the numerous clinical associations with human par-
The patient reported by Kaiser and Bradford581 is vovirus B19 infection.593 Indeed, AIHA is not even
unique in that the patient developed paroxysmal cold mentioned in some extensive reviews of the clinical
hemoglobinuria during the prodromal stage of manifestations of infection with this virus.90,594
chickenpox. Nevertheless, there have been a few case reports sug-
Ziebold and coworkers583 conducted a survey of gesting that infection with parvovirus B19 may have
severe complications in immunologically normal chil- resulted in AIHA.29,82,174,595,596
dren with varicella throughout Germany during a 1- Bertrand and coworkers82 described a 12-year-old
year period. They estimated that the incidence of boy who was admitted to the hospital with pancy-
severe chickenpox complications was 8.5:100,000 chil- topenia and an erythroblastopenic marrow with
dren but found only 6 of 119 reported severe compli- normal granulocytic maturation and rare megakaryo-
cations consisted of hematologic abnormalities. Of cytes. He also had a positive DAT. The patient was
these, 5 children had thrombocytopenia and one child transfused and the thrombocytopenia and neutrope-
developed anemia and neutropenia that lasted for at nia spontaneously resolved within 3 days. However,
least 3 months. However, the anemia was not further the positive DAT persisted and he developed a high
characterized. reticulocyte count and bilirubinemia. The authors
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 107

TABLE 3-25. REPORTS OF COLD AGGLUTININS IN RUBELLA INFECTION

Rubella Cold
Serologically Agglutinin Immunoglobulin Titer Clinical
Patient Age Ascertained Specificity Class and Type (4°C) Manifestation

1 19 yr Yes Anti-Pr3 IgM κ 8000 Acrocyanosis


2 Newborn No Anti-Pra IgM κ 512 None
3 Unknown No Anti-Pr1 IgM κ 128 —*
4 Newborn Yes Anti-Pra IgM κ 64 None
5 Newborn Yes Anti-Pr1 IgM λ 16 None
6 5 yr Yes — — — Acrocyanosis
7 5 yr Yes Anti-Pr1 IgG λ 512 Hemolytic anemia

* Undetermined.
Modified from Konig AL, Schabel A, Sugg U, Brand U, Roelcke D: Autoimmune hemolytic anemia caused by IgG lambda-monotypic cold agglutinins
of anti-Pr specificity after rubella infection. Transfusion 2001;41:488–492.

speculated that the patients’ anemia was caused by a Hepatitis. Hemolytic anemia in patients with liver
double mechanism related to the parovovirus infec- disease may be caused by a variety of mechanisms of
tion: anti-erythrocyte autoantibody formation and which AIHA is among the most unusual.597 Although
erythroblastopenia. The latter resulted in the patient’s there are a number of reports of AIHA in association
severe anemia, which would not have developed so with hepatitis, the immune causation of the hemolysis
acutely if not for the hemolysis. Thus, the AIHA was is often poorly documented and, in a number of
revealed by the parvovirus linked erythroblastopenia. instances, associated immune deficiency disorders or
Kunimi and coworkers595 reported two patients drug therapy may be more significant than the pres-
with AIHA associated with parvovirus infection. Both ence of the hepatitis. Further, the association of the
patients presented with manifestations of AIHA and two disorders may merely be coincidental since hepa-
tests for parvovirus infection were positive. However, titis of a variety of etiologies is so common.
the authors pointed out that the parvovirus infection AIHA in association with hepatitis C virus (HCV)
and AIHA could have occurred independently. infection in patients without an underlying immune
de la Rubia and coworkers596a reported a 5-year-old deficiency or therapy with drugs known to cause
girl with homozygous α-thalassemia who had a tran- hemolysis is rare. Nevertheless, Ramos-Casals and
sient aplastic crisis due to B19 infection. She recovered coworkers597a reported 35 cases of severe autoimmune
and was discharged with a hemoglobin near her basal cytopenias in treatment-naïve hepatitis C infection,
value (7.4 g/dL). However, four days later she was including 17 patients with AIHA. A diagnosis of AIHA
admitted with a hemoglobin of 4.4 g/dL and manifes- was made on the basis of a decrease of at least 2 g/dL
tations of AIHA including a positive DAT (C3d) and a in hemoglobin levels, an increase of at least 0.6 mg/dL
positive antibody screening test due to a high titer in the serum unconjugated bilirubin level, a reticulo-
cold agglutinin (titer 1024 at 4°C and 64 at 30°C). She cyte count >5%, and a positive DAT. However, they
was treated with prednisone, IVIG, RBC transfusions, did not provide data regarding the presence of RBC
and plasma exchanges, and her clinical course gradu- autoantibodies, and 12 (71%) of the patients had associ-
ally improved with the hemoglobin level returning to ated autoimmune diseases.
baseline values (7 to 8 g/dL). The authors suggested Chao and coworkers598 described a 69-year-old
that parvovirus B19 infection should be sought in woman with HCV-related liver cirrhosis who
patients with AIHA, particularly when associated developed anemia, reticulocytosis, a positive DAT
with reticulocytopenia. Likewise, B19 should be listed and IAT, and IgG warm autoantibodies. After pred-
among the viral illnesses associated with acute AIHA. nisolone therapy, her anemia improved dramatically.
Chambers and coworkers174 reported a 3-year-old Similarly, Srinivasan599 reported a patient with HCV
boy who had a febrile illness with rash typical of acute infection who developed hemolytic anemia with a
parvovirus (fifth disease). The hematocrit was 14.5%, positive DAT. Prednisone therapy resulted in resolu-
haptoglobin was undetectable, LDH and bilirubin tion of the hemolysis and did not result in a deterio-
were elevated, but the reticulocyte count was only ration of the underlying HCV liver disease, although
2.3%. The DAT was positive with anti-C3 and anti-IgG an increase in HCV viremia was noted. Previous
reagents, and both IgM and IgG antibody against par- reports of HCV infection and AIHA included patients
vovirus were detectable, confirming recent infection. with an associated immune deficiency disorder600 or
The Donath-Landsteiner test was positive. The patient interferon therapy,601-604 although Moccia and
was treated with RBC transfusion, corticosteroids, and coworkers605 reported AIHA in a patient with chronic
IVIG, and his anemia rapidly resolved. The authors hepatitis C who had never been treated with inter-
concluded that parvovirus should be included among feron. Also, Longo and coworkers606 reported a
infections which antedate AIHA in children and that patient who developed acute hepatitis and AIHA,
the unexpected low reticulocyte count was probably a both of which were attributed to treatment with oral
result of the parvovirus infection. ceftriaxone.
108 Immune Hemolytic Anemias

Serologic findings in purported cases of AIHA asso- patients. Jeje and coworkers644 measured RBC-
ciated with hepatitis are often not definitive, making associated IgG on washed red cells from 26 children
uncertain the immune cause of the hemolysis. Kondo with Plasmodium falciparum parasitemia using an
and coworkers607 reported a patient with autoim- immunoradiometric assay. The amount of RBC-
mune hepatitis who developed hemolysis but had a associated IgG was related to the hemoglobin
negative DAT. Nevertheless, elevated levels of RBC- concentration, degree of parasitemia, and serum immu-
associated IgG were detected and these decreased as noglobulin level. The mean RBC-associated immuno-
the patient improved after corticosteroid therapy. globulin for patients with malaria was 629 molecules
Other patients with DAT-negative hemolysis were per red cell (range, 215 to 1770), which was significantly
reported in association with hepatitis A608 and chronic higher than the control group, who had a mean of 395
hepatitis C.604 molecules per red cell (range, 190 to 930). A statistically
Giant cell hepatitis with AIHA has been reported to significant correlation was established between RBC-
be a distinct entity with an aggressive course.609-614 associated IgG and the degree of anemia in the infected
Early and sustained immunosuppression has been patients. They suggested that their observations
recommended to control the progressive hepatocellu- supported the hypothesis than an immunologic mecha-
lar damage and to prevent cirrhosis. Hadzic and nism could be involved in the anemia associated with
coworkers615 described a patient who was diagnosed malarial infections.
with Evans’s syndrome at 8 months of age and at the Berzins and coworkers645 studied the specificity of
age of 5 years developed liver dysfunction. Liver his- RBC antibodies in the serum of patients with malaria
tology revealed severe giant cell transformation with and found that most malarial sera showed elevated
colestasis, marked inflammation, clusters of neu- levels of antibodies against several RBC polypeptides.
trophils, spotty hepatocyte necrosis, and advanced Wahlgren and coworkers646,647 characterized the
fibrosis. She responded poorly to therapy and died 11 humoral immune response in Plasmodium falciparum
months later. The authors suggested that the occur- malaria. They tested 175 sera for their reactivity with
rence of Evans’s syndrome and giant cell hepatitis in antigens derived from late trophozoites or schizonts
their patient were not coincidental, but suggestive of a from P. falciparum. They found that patients had anti-
common immune dysregulation mechanism. bodies that reacted with the antigens, and that they
There have been isolated reports of AIHA in had a high parasite specificity as indicated by inhibi-
patients with other forms of hepatitis, including tion and absorption experiments. Many sera also had
autoimmune hepatitis,607,616 primary biliary cirrh- elevated levels of antibodies specific for RBC antigens.
osis,2,222,617-620 necrotic hepatitis and peliosis,621 A correlation, most pronounced in the IgM system,
infection with hepatitis B virus,622,623 hepatitis A was also seen between the anti-RBC and the anti−P.
virus,74,608,624 and chronic active hepatitis.625-630 falciparum antibody levels.
Infection with Malaria and Other Blood Parasites. Topley and coworkers648 found that 13 of 23 patients
As early as 1908, Christophers and Bently631 described had a positive DAT with an anti−non-gamma globulin
the engulfment of large numbers of apparently reagent. Rosenberg and coworkers649 reported 56
normal RBCs by splenic phagocytes in patients with patients who had a significant rise in serum IgM levels
malaria. Indeed, the extensive destruction of red cells and reduced C3 levels; 52 had IgM and 31 had IgG on
by some mechanism other than direct parasitic attack their RBCs as demonstrated by a fluorescent antibody
has been recognized for about a century,632 and technique. Facer and coworkers635 examined 160 chil-
numerous investigators have reviewed the evidence dren and found that 50% of them had a positive DAT,
suggesting an immune mechanism as a contributing usually due to the presence of C3d or IgG and C3d on
factor in the hemolysis.632-642 their RBCs. Abdalla and coworkers638 found that 16 of
Data Indicating Mechanisms of Anemia Other than 17 patients tested had a positive DAT with a wide spec-
Direct Rupture of Parasitized RBCs. A number of trum antiglobulin reagent and 9 also reacted with anti-
findings clearly indicate that the anemia of malaria is IgG sera. Woodruff and coworkers634 found that RBCs
not completely explained by rupture of parasitized were coated with complement in 3 of 5 patients with
RBCs.633,642 (1) The degree of anemia in malaria is P. falciparum infections.
not correlated with the degree of parasitemia.632 Wenisch and coworkers650 pointed out that changes
(2) Hemolysis can appear or persist after the infection in the complement system have long been described
is cured and no more parasites can been seen in the in malaria. Components of the classical pathway have
peripheral blood.632,634,643 (3) Phaagocytosis of non- been demonstrated to be depressed in malarial infec-
parasitized RBCs has been documented.632,633,641 tion, whereas components of the alternative pathway
Ineffective erythropoiesis with dyserythropoiesis may have been reported to be largely unaffected. In
be a prominent finding638; and inhibition of red cell contrast, Wenisch and coworkers650 found that both
production is commonly present.634,638 the classical and alternative pathways of the comple-
Evidence Suggesting an Immune Etiology as a ment system are profoundly activated in complicated
Contributing Factor to the Hemolysis of Malaria. There malaria. However, no relation between clinical disease
are numerous suggestions that immune mechanisms severity and complement fragments existed.
play a role in the causation of anemia in malaria. IgG, Facer and coworkers636 reported that eluates form
IgM, or complement has been found on the RBCs of the RBCs of Gambian children with P. falciparum infec-
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 109

tion had specific antibody activity against P. falciparum was 89.6 days compared with 56.8 days for autologous
schizont antigen as demonstrated by means of the erythrocytes. However, the authors found no increase
indirect fluorescent antibody technique. Cross-reac- in cell-bound IgG or C3 and they concluded that the
tion was not observed with either P. falciparum game- increased rate of RBC destruction is not antibody or
tocytes of P. malariae schizonts. No blood group complement mediated.
specificity could be demonstrated in either the RBC In instances in which an immune pathogenesis has
eluates or in serum taken from children with a posi- been suggested, the most commonly proposed mech-
tive DAT. These authors suggested that parasite anisms include antibodies reactive against malarial
antigen-antibody immune complexes likely con- antigens adsorbed to RBCs, immune complexes
tribute to the pathogenesis of the anemia in falciparum adsorbed to RBCs, or antibodies against RBC antigens
malaria.635 exposed by malarial parasites. These mechanisms
Lefrancois and coworkers651 described 14 patients would not represent true autoimmune reactions but
with chronic falciparum malaria who had cold antibod- instead would be examples of bystander immune
ies of anti-I specificity in their serum. In five the DAT hemolysis,655-657 a topic that is reviewed in Chapter 9.
was positive and in the other nine patients autoanti- Other Parasitic Diseases. The mechanism whereby
bodies were revealed by elution. However, the titers of anemia is produced in kala-azar (systemic leishmania-
the antibodies were tested only at 4°C and only two of sis) was studied first among the parasitic diseases,
the 14 had titers as high as 128 at that temperature. because the causal parasite lives solely outside erythro-
Drouin and coworkers639 reported a 30-year-old cytes, yet the untreated infection is associated with
woman who contracted infection with P. falciparum severe anemia.634 The question was whether this
during the first trimester of her pregnancy. Her illness anemia results from hemolysis, marrow suppression,
was characterized by hemolytic anemia with a or a combination of both. 51Cr and 59Fe studies indi-
strongly positive DAT using anti-IgG, and negative cated that hemolysis was mainly responsible.634
reactions using anti-C3d, -C3b, and -C4. Eluates from Kokkini and coworkers658 reported a 3-year-old boy
the patient’s erythrocytes contained IgG1 antibodies with systemic leishmaniasis who developed CAS. The
that reacted strongly with RBC panels in a nonspecific DAT was positive using a polyspecific reagent and
pattern consistent with results found in WAIHA. Treat- with anti-C3b, -C3d, and -C4, but negative using anti-
ment of the infection led to resolution of the hemolysis. IgG. Studies of reactivity of the patient’s serum
Ritter and coworkers652 found IgM antibodies revealed a cold agglutinin of anti-I specificity. At 4°C
specific for the glycolytic enzyme triosphosphate iso- the antibody reacted to a titer of 16,384 against normal
merase in the serum of 4 patients with P. falciparum adult RBCs, 8192 against the patient’s own RBCs,
infection who had prolonged hemolysis after clear- 32,768 against enzyme-treated RBCs, and 312 against
ance of parasites from their blood. In vitro, affinity cord RBCs. The antibody agglutinated normal adult
purified antibodies induced lysis of normal RBCs and RBCs up to 25°C and the enzyme-treated RBCs up to
activation of complement. The antibodies were only 36°C. The patient required transfusion of 6 units of
detected in the 4 patients who had prolonged hemol- RBCs during the first 47 days of hospitalization but,
ysis and anemia and not in patients with uncompli- after treatment for the underlying infection, hemoglo-
cated courses of P. falciparum or P. vivax malaria. The bin levels became normal, and 31⁄2 months later no cold
DAT was negative in all patients. The authors sug- agglutinins were present and the DAT was negative.
gested that autoimmune mechanisms have an impor- Pontes de Carvalho and coworkers659 studied 34
tant role in the development of anemia in malaria. patients with American visceral leishmaniasis and
Conclusions. It is clear that anemia associated with found IgG molecules on RBC membranes in all of
malaria has a multifactorial pathogenesis including them. They determined that the RBC-bound IgG was
mechanical rupture of RBCs by the intracellular para- not RBC-specific autoantibody, but that that the IgG
site, possible immune destruction of RBCs, increased had anti-Leishmania activity. They concluded that the
erythrophagocytosis, decreased erythropoiesis, and IgG may be a component of Leishmania antigens−anti-
sequestration of parasitized and normal RBCs.633 Leishmania immune complexes and that the patho-
Numerous authors have suggested that immune genesis of anemia in the disease was multifactorial.
mechanisms, including autoimmunization, contribute Hemolytic mechanisms were also found to be an
to the pathogenesis of the anemia. Although such data important factor in the production of anemia in
are suggestive, compelling evidence is lacking and African trypanosomiasis, which is another disorder in
other investigators have found no evidence for an which the parasite remains solely outside of the
immune mechanism,643,653,654 or have suggested that erythrocyte.634 Kobayakawa and coworkers660
autoantibodies were not pathogenic.633 Looareesuwan studied antibody production by the spleen in a
and coworkers643 studied RBC survival times in murine model and observed autoimmune responses
healthy Thai controls and in patients after clearance of to DNA, RBCs, and thymocyte antigens in mice
asexual P. falciparum or P. vivax parasitemia. In P. vivax infected with Trypanosoma brucei.
malaria the mean cell life of autologous RBCs in seven Wolf and coworkers661 reported that 3 of 30 patients
patients was a 67.2 days and that of compatible donor with clinical and laboratory evidence of babesiosis had a
cells in six patients was 66.8 days. In 12 P. falciparum positive DAT, with noncomplement-binding IgG anti-
patients the mean cell life of compatible donor RBCs bodies coating their RBCs. The autoantibodies were
110 Immune Hemolytic Anemias

nonreactive, or markedly less reactive, with Rhnull asitic antigens involved in the immune complex for-
cells; and eluates prepared from the patients’ RBCs mation were unsuccessful and there was not a good
reacted with RBCs taken from healthy individuals. correlation with hemagglutination titers.
They suggested that the autoimmune response is Bacterial Infections. Bacterial infections have not
aimed at integral structures of the RBC membrane, been reported frequently as the cause of AIHA except
possibly associated with Rh protein, and does not for the association of Mycoplasma pneumoniae infection
appear to be directed against unique antigens of para- with CAS. There are, however, a few reports of AIHA
sitic origin. The role of the autoantibodies in the in association with other bacterial infections including
pathogenesis of the hemolytic anemia is difficult to Klebsiella pneumonia, pneumococcal bacteremia,
determine, since only 10% of the patients studied by Hemophilus influenzae type b, Yersinia enterocolitica,672
Wolf and coworkers661 had a positive DAT, and and Legionella pneumophila.673 Although hemolysis is
hemolysis is an intregral part of the clinical spectrum generally well documented in such reports, autoanti-
of infection with babesiosis.662,663 bodies directed against intrinsic RBC antigens are
Bowsher and coworkers664 reported a child with generally not present so that their designation as cases
leptospirosis who had pneumonia, pleural effusion, of AIHA is not definitive.
and anti-I cold hemagglutinin antibodies. The clinical Several authors have noted the hemolytic nature
presentation of the patient also included hemolytic of anemia associated with infections caused by
anemia. Unfortunately, no characterization of the anti- H. influenzae type b, and have suggested the possibility
I was performed so that its possible role in the that adsorbed bacterial toxins contribute to RBC
patient’s hemolytic anemia cannot be ascertained. destruction.674-677 Shurin and coworkers674 found that
Van Audenhove and coworkers665 reported a the capsular polysaccharide of this organism, polyribo-
patient with Bartonella henselae infection who had syl ribitol phosphate (PRP), binds to human RBCs in
hemolytic anemia. The DAT and IAT were negative as vivo and that PRP-coated RBCs have a shortened cir-
were tests for cold agglutinins and Donath- culation time in mice. The authors demonstrated that
Landsteiner antibodies. The authors concluded that antiserum to PRP induces intravascular, complement-
this was a case of DAT-negative AIHA triggered by mediated hemolysis, as well as extravascular hemoly-
the parasitic infection, although they presented no sis, and reported that patients with invasive infection
data to substantiate this supposition. develop hemolysis when circulating PRP and antibody
Giirelli and coworkers666 reported a 73-year-old man to PRP are present simultaneously.
with hydatid disease who developed AIHA due to an Caceres678 described a case of WAIHA associated
IgM cold agglutinin with anti-I specificity. The patient’s with pneumococcal sepsis. A 57-year-old man was
hemoglobin was 3.5 g/dL, and signs of hemolysis admitted with high fever (40°C) and a left lower bron-
included hemoglobinuria, total serum bilirubin of chopneumonia. The hemoglobin was 5.4 g/dL, and
4.4 mg/dL with indirect bilirubin 3.3 mg/dL, and a reticulocytes 33.7%; the DAT and IAT were positive
bone marrow that showed erythroid hyperplasia. The and the patient’s RBCs were coated with IgG and C3d.
DAT was positive with polyspecific and anticomple- Blood cultures and a sputum culture were positive for
ment reagents. An agglutinin acting against normal growth of Streptococcus pneumoniae. The patient recov-
adult cells and the patient’s RBCs (1:128) and cord cells ered rapidly after treatment with penicillin, intra-
(1:4) was found in the patient’s serum at 4°C. The anti- venous gammaglobulins, methylprednisolone, and
body reacted up to 32°C against autologous and allo- blood transfusion.
geneic adult RBCs. 2-Mercaptoethanol treatment As described previously (page 74), a number of
abolished the agglutinating activity. Hemolysis did not reports have indicated a frequent association of PCH
improve with corticosteroid treatment, but specific and infection. Although the infections are most fre-
therapy for hydatid disease (mebendazole) caused a quently viral, infections with H. influenzae,175 M. pneu-
remission of that disorder, normalization of his hemo- moniae,179,180 and Klebsiella pneumonia181 have also
globin level, and resolution of all immunohematologic been implicated.
abnormalities. The authors investigated 44 additional
patients with hydatid disease and found only 1 with an PRIMARY IMMUNODEFICIENCY DISEASES
IgM cold autoantibody, but with no signs of anemia.
Several groups have demonstrated that anti-I are The primary immunodeficiency diseases are charac-
inhibited by hydatid cyst fluid,667-669 suggesting the terized by lifelong susceptibility to chronic or recur-
possibility that the stimulation of the host immune rent infections, cancers (especially lymphomas), and,
system could occur if antigenic material from cysts in some cases, unregulated inflammation due to
were to be released into the bloodstream. Also, abnormal infiltration of lymphocytes in tissues and
Perricone and coworkers670 found that hydatid cyst organs.678a Another common manifestation of primary
fluid activated human complement by the alternative immunodeficiency is autoimmune disease.600,679-686
and the classical pathways. D’Amelio and cowork- The most common autoiommune disorder associated
ers671 tested sera from eight patients with dissemi- with immunodeficiency diseases is immune ITP, fol-
nated or localized hydatid disease for the presence of lowed by AIHA.679 The congenital immunodeficiency
circulating immune complexes. Efforts to identify par- conditions that are most associated with autoimmune
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 111

TABLE 3-26. CONGENITAL IMMUNODEFICIENCY Oyefara and coworkers689 reported three patients
DISEASE AND AUTOIMMUNE SYNDROMES who developed RBC autoantibodies, two of whom had
AIHA, in association with chronic mucocutaneous can-
Immunodeficiency Disease Autoimmune Syndrome
didiasis, which is an immunodeficiency disease charac-
terized by T-cell dysregulation and chronic superficial
IgA deficiency and common ITP candidal infections. They suggested that the RBC auto-
variable immunodeficiency Autoimmune hemolytic anemia antibodies in these cases may be related to immuno-
(AIHA) regulatory disorders in this disease, and that all
Rheumatoid arthritis
Pernicious anemia
patients with the mucocutaneous candidiasis should be
Juvenile rheumatoid arthritis screened periodically for RBC autoantibodies.
Hyper-IgM syndrome Neutropenia The Wiskott-Aldrich syndrome is an X-linked dis-
ITP order characterized by eczema, thrombocytopenia,
AIHA and recurrent infection. A DAT-positive hemolytic
Inherited defects of complement SLE anemia has been reported in 5% to 10% of patients
Vasculitis with the syndrome.690-693
Glomerulonephritis The DiGeorge anomaly is a well-recognized spec-
Henoch-Schonlein purpura
trum of disease syndromes consequent on abnormal
Autoimmune lymphoproliferative ITP
disease AIHA
development of the third and fourth pharyngeal
Neutropenia pouches, and is associated with varying degrees of
Mucocutaneous candidiasis Diabetes thymic hypofunction. The immunodeficiency of the
Hypoadrenalism defect is variable. Pinchas-Hamiel and coworkers694
AIHA reported a patient with DiGeorge syndrome who
Wiskott-Aldrich syndrome Juvenile rheumatoid arthritis developed AIHA, thrombocytopenia, and chronic
AIHA persistent hepatitis.
Modified from Cunningham-Rundles C: Hematologic complications of primary
Although the association between cytopenias and
immune deficiencies. Blood Rev 2002;16:61–64. congenital immune deficiency is unclear, defects in T-
cell regulation, cytokine defects, abnormal apoptosis,
and abnormal production of immunoglobulins with
autoimmune features are potential mechanisms.679
AIHA in Patients with Severe Combined
conditions are listed in Table 3-26. Selective IgA Immunodeficiency (SCID) after T-Cell–Depleted
deficiency and common variable immunodeficiency Hematopietic Cell Transplantation. Horn and cowork-
(CVID) are among the most common congenital ers685 reported that 8 of 41 patients (19.5%) with SCID
immunodeficiencies with frequencies of 1:500 and who underwent a T-cell−depleted hematopoietic cell
1:20,000, respectively.679 transplant developed AIHA. The authors commented
Hansen and coworkers687 reported a 26-year-old that AIHA was more common than GVHD in this
male with a 10-year history of selective IgA deficiency group of patients and it contributed significantly to
and recurrent Evans’s syndrome. Both serum IgA and morbidity and mortality. Other than infections, AIHA
saliva secretory IgA were below the detection limit
(<0.05 mg/L). The patient had no other clinical
manifestations of autoimmune disease, and the authors
emphasized the pleomorphic and randomly appearing TABLE 3-27. AUTOIMMUNE DISEASE IN 248
immunologic features of selective IgA deficiency. PATIENTS WITH CVID
CVID is characterized by reduced serum
Condition Males (n) Females (n)
immunoglobulins and heterogeneous clinical features,
and is usually diagnosed in individuals between the ITP 9 9
ages of 20 and 40 years. Even though serum immu- Hemolytic anemia 6 6
noglobulins may be quite reduced in CVID, about 23% Rheumatoid arthritis 0 5
of subjects develop autoimmune disease, with ITP and Juvenile rheumatoid arthritis 2 2
Anti-IgA antibody 1 6
AIHA among the most prevalent (Table 3-27). In a Sicca syndrome 0 2
series of 248 patients with CVID, there were 12 cases of Primary biliary cirrhosis 1 2
AIHA.688 Of these, four had concurrent granuloma- Alopecia totalis 2 2
tous disease and three had been previously diagnosed Pernicious anemia 0 3
Hyperthryroidism 0 2
with sarcoidosis. Five patients had prolonged medical Autoimmune neutropenia 1 1
treatment with corticosteroids alone or in combination Nephrotic syndrome 2 0
with IVIG (1 to 2 g/kg) with no apparent benefit, and SLE 1 1
ultimately had to undergo splenectomy, which was Vasculitis 2 1
successful in 11 of the 12 cases. Postoperative com- Totals 27 42
plications occurring after splenectomy included From Cunningham-Rundles C: Hematologic complications of primary immune
cutaneous fistulae and sepsis in three cases. deficiencies. Blood Rev 2002;16:61–64.
112 Immune Hemolytic Anemias

was the most common post-transplant complication in


this patient cohort. In contrast, the incidence of AIHA TABLE 3-28. RESULTS OF TESTING FOR IgG
among the authors’ patients who underwent T-cell− AND COMPLEMENT ON RBCS OF 21 DAT-POSITIVE
depleted marrow transplantation for disorders other ALPS PATIENTS
than SCID was only 6%.
History of No History of
The AIHA developed at a median of 8 months after Hemolytic Anemia Hemolytic Anemia
transplantation. All patients had warm-reacting Reactive Reagent (n = 10) (n = 11)
autoantibodies, and two of the eight had concurrent
cold and warm autoantibodies. The clinical course IgG only 6 (60%) 3 (27%)
was most severe in the two patients who had cold and Complement only 0 (0%) 1 (9%)
IgG plus complement 3 (30%) 1 (9%)
warm autoantibodies. Five patients received specific Polyspecific only 1 (10%) 6 (55%)
therapy for AIHA. Immunosuppressive therapy with
cyclosporine and steroids was generally effective. From Stroncek DF, Carter LB, Procter JL, Dale JK, Straus SE: RBC autoantibodies
in autoimmune lymphoproliferative syndrome. Transfusion 2001;41:18–23.
However, treatment of the two patients with concur-
rent cold-reacting autoantibodies and other immune
cytopenias had a severe clinical course and required ALPS.703,704 The genetic mutations that cause ALPS
additional treatments. In these patients treatment of are inherited in an autosomal dominant manner, but
the AIHA was lengthy, and successful taper of immu- clinical penetrance of the disease is variable.
nosuppression was only possible after achieving RBC Autoantibodies in ALPS. Stroncek and
T-cell reconstitution. coworkers698 studied 34 consecutive patients with
The authors hypothesized that development of ALPS and 37 of their relatives without ALPS. The
AIHA in these cases may be related to a paucity of DAT was positive in 21 of the 34 ALPS patients
regulatory T cells, which may result in the emergence (61.8%) (Table 3-28) and in one (3%) of the unaffected
of unregulated immunoglobulin-producing B-cell relatives (P = 0.001). Antibodies were eluted from the
clones, and delayed T-cell reconstitution may account RBCs of 9 of the 21 patients with a positive DAT. In
for their persistence. Indeed, after reconstitution of eight patients, the eluted antibody was found to be a
T-cell immunity, a reduction of immunosuppressive panagglutinin, and RBC antibodies were detected in
therapy for AIHA was possible. the serum of seven ALPS patients by IAT. Ten of the 21
Other authors have reported rather similar data. ALPS patients with a positive DAT had a history of
Drobyski and coworkers695 described AIHA in 5% of hemolytic anemia. Thus, approximately two thirds of
patients who survived at least 6 months following T- ALPS patients have positive DATs with panaggluti-
cell−depleted marrow transplantation, and Godder and nating IgG autoantibodies coating their RBCs. One
coworkers696 described AIHA in 7.5% of patients receiv- third of these patients had a history of WAIHA. No
ing a T-cell−depleted haploidentical marrow transplant. other syndrome is associated with such a high inci-
dence of RBC autoantibodies and AIHA.
AUTOIMMUNE LYMPHOPROLIFERATIVE The proposed mechanism of autoantibody forma-
SYNDROME tion in ALPS is unregulated B-cell activation and/or
accumulation of autoreactive B cells due to a defect in
Autoimmune lymphoproliferative syndrome (ALPS), Fas-mediated lymphocyte apoptosis.700
also referred to as Canale-Smith syndrome,697 is Eleven of 21 ALPS patients with a positive DAT had
a genetic disease that presents in early childhood a nonreactive eluate and no history of hemolytic
and is associated with abnormal lymphocyte apopto- anemia (see Table 3-28). These DATs are probably
sis, chronic nonmalignant lymphadenopathy, and related to the fact that serum IgG levels are elevated in
splenomegaly.698-703 A subset of rare peripheral blood ALPS patients, as it is known that elevated IgG levels
and tissue lymphocytes that express α and β T-cell are associated with positive DATs.705 In determining if
receptors, but not CD4 or CD8 antigens (TCR α/β+ relatives of ALPS patients are clinically and geneti-
CD4− CD8− cells, or so-called “double-negative” cally affected, Stroncek and coworkers698 found that
T cells) are expanded. Patients with ALPS commonly the DAT was the most useful serologic marker.
exhibit episodic and intermittent severe AIHA, and When a diagnosis of WAIHA is made, one must be
autoimmune thrombocytopenia, neutropenia, or aware of the possibility of ALPS, especially in children
combined cytopenias.700 and in cases of familial AIHA.
In many ALPS patients, defects in receptors and
enzymes involved with lymphocyte apoptosis have MISCELLANEOUS DISORDERS REPORTED
been identified. The most common mutations involve IN ASSOCIATION WITH AIHA
APT1, which encodes for Fas receptor, also known as
APO-1 or CD95. Patients with both ALPS and APT1 Occasional cases of AIHA have been reported in
mutations have been characterized as having type Ia association with a wide variety of disorders including
ALPS; patients with defects in the Fas ligand have B-cell acute lymphocytic leukemia,706 leukemia
type Ib ALPS, patients with defects in caspase of large granular lymphocytes,707 hairy-cell leuke-
enzymes have type II ALPS, and patients in whom the mia,354,708 acute myelocytic leukemia,709 chronic
defect in apoptosis has yet to be defined have type III myelogenous leukemia,710-712 angioimmunoblastic
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 113

lymphadenopathy with dysproteinemia (AILD),713-718


TABLE 3-29. PREVALENCE OF COLD AGGLUTININS
myelodysplastic syndromes,209,719 sideroblastic
IN DISEASES WITH IgM M-COMPONENTS
anemia,720 Castleman’s disease,721-724 multiple mye-
loma,2,10,725-729 cutaneous T-cell lymphomas,730-733
Percent
thrombotic thrombocytopenic purpura,734 Sjögren’s No. of No. with with Cold
syndrome,735-742 disseminated encephalomyelitis,743 Diagnosis Cases Cold Agglutinins Agglutinins
thyroid disease,744-751 Kawasaki disease,752-756 sar-
coidosis,757-761 thalassemia,762-767 and agnogenic Lymphoma 63 17 27
myeloid metaplasia.2,768,769 Leukemia 30 5 17
Waldenström’s 105 13 12
macroglobulinemia
Secondary Cold Agglutinin Syndrome Cancer 26 0 0
Collagen vascular 21 0 0
As indicated earlier, cold agglutinin disease consti- diseases
tutes a minority of cases of cases of secondary AIHA. Miscellaneous* 63 8 13
Total 308 43 14
Some associated disorders merit special comment.
* Includes benign monoclonal gammopathy, chronic liver diseases, IgM
myeloma; amyloidosis, and six cases of chronic cold agglutinin disease.
INFECTIOUS DISEASES From Crisp D, Prazanski W: B-cell neoplasms with homogeneous cold-reacting
antibodies (cold agglutinins). Am J Med 1982;72:915–922.
M. pneumoniae infection and infectious mononucleosis
are the most common infectious diseases associated
with CAS. Less commonly, CAS may be associated
with rubella, varicella, CMV, and HIV infection, as between 26% and 33% in 3 cases, and between 41% and
reviewed earlier. In addition, King and May673 95% in 6 cases. In several patients, the lymphoid cells
reported CAS in a patient with Legionnaires’ disease. increased during the course of the disease. On the basis
of such findings some authors feel that chronic CAS
WALDENSTRÖM’S MACROGLOBULINEMIA is probably a variant of Waldenström’s macroglo-
bulinemia771 in which the IgM M-component has cold
A number of authors have noted similarities of labora- agglutinin activity.103,116,117
tory findings in patients with CAS and patients who Crisp and Pruzanski107 reviewed findings in
have monoclonal IgM proteins without cold agglutinin 308 patients with monolonal macroglobulins inves-
activity. In both instances, bone marrow findings may tigated in their laboratory and reported that 43
consist of increased numbers of abnormal lymphoid (14%) had cold agglutinin activity (Table 3-29). In 37 of
and plasma cells.770 Schubothe103 performed serial these 43 patients, detailed clinical and laboratory data
examinations of the bone marrow with quantitative dif- were available and, in addition, they reviewed 41 previ-
ferential counts in 14 patients with CAS. The number of ously reported patients with “persistent” cold agglu-
lymphoid cells was between 10% and 24% in 5 cases, tinins. The most common diagnoses in these

TABLE 3-30. CLINICAL MANIFESTATIONS OF B-CELL NEOPLASMS WITH COLD AGGLUTININS

Lymphoma Waldenström’s Chronic Lymphocytic Chronic Cold


Disease Macroglobulinemia Leukemia‡ Agglutinin

No. of patients 31 13 6 28
Age range (mean) (yr) 38–80 (63.2) 18–86 (61.3) 43–87 (63.3) 17–95 (61.2)
M/F 14/17 9/4 2/4 13/15
Clinical manifestations (n)*
Fatigue, weakness 15 (48) 6 (48) 3 12 (43)
Fever 8 (26) 0 0 3 (11)
Raynaud’s phenomena 5 (16) 3 (23) 2 10 (36)
Hemolytic crises 3 (10) 1 (8) 1 12 (43)
Bleeding phenomena 5 (16) 4 (31) 0 15 (54)
Lymphadenopathy 15 (48) 3 (23) 3 2 (7)
Hepatomegaly 14 (45) 8 (62) 2 4 (14)
Splenomegaly 22 (71) 7 (54) 4 10 (36)
Survival time (mo)†
Start to death 36 [11] 65.6 [3] – 42.3 [3]
Diagnosis to death 20.1 [19] 29.6 [7] – 36.8 [4]
Start to last follow-up 12 [3] 49.0 [2] 105 [4] 80.7 [14]
Diagnosis to last follow-up 25.4 [7] 32.7 [3] 81 [4] 65.0 [20]

* Percentage given in parentheses.


† Number of patients given in brackets.
‡ Due to a small number of patients the percentages are not presented.
Modified from Crisp D, Pruzanski W: B-cell neoplasms with homogeneous cold-reacting antibodies (cold agglutinins). Am J Med
1982;72:915–922.
114 Immune Hemolytic Anemias

TABLE 3-31. LABORATORY DATA IN B-CELL NEOPLASMS WITH COLD AGGLUTININS

Chronic Chronic-Cold
Waldenström’s Lymphocytic Agglutinins
Data Lymphoma Macroglobulinemla Leukemia Disease

Hemoglobin (g/dL)
<8.5 19 (66) 7 (58) 2 13 (47)
8.5–10.4 5 (17) 2 (17) 1 6 (21)
>10.4 5 (17) 3 (25) 2 9 (32)
Reticulocytosis (≥2%) 16/31 (52)* 8/8 (100) 5/5 19/22 (86)
Positive Coombs’ test results
Direct 17/23 (74) 5/6 (83) 3/3 12/19 (63)
Indirect 5/8 (63) 0/3 (0) 1/1 4/5 (80)
Lymphocytosis (≥4000/mm3) 6/24 (24) 4/9 (44) 6/6 5/24 (21)
Cryoglobulinemia 7/19 (37) 2/6 (33) 2/5 2/21 (10)
Cold agglutinins
Anti-I 8 (32) 3 (33) 2 17 (24)
Anti-i 9 (36) 3 (33) 1 4 (17)
Other 8 (32) 3 (33) 0 2 (9)
Not defined 6 4 3 5
Light chains
κ 7 (29) 5 (71) 3 11 (92)
λ 17 (71) 2 (29) 0 1 (8)
Unknown or not reported 7 6 3 16
Immunoglobulins
IgM >200 mg/dL 24/27 (89) 7/7 (100) 2/3 20/22 (91)
Low IgG and/or IgA (<600 mg/dL and/or 11/27 (41) 2/7 (29) 3/3 10/19 (53)
<100 mg/dL)
Bence Jones proteinuria 8/13 (62) 4/5 (80) 2/2 8/11 (73)

* Number of patients with positive findings/number of patients investigated; percentage in parentheses.


From Crisp D, Pruzanki W: B-cell neoplasms with homogeneous cold-reacting antibodies (cold agglutinins). Am J Med 1982;72:915–922.

78 patients were chronic cold agglutinin disease, defined as a lymphoplasmacytic lymphoma character-
lymphoma, chronic lymphocytic leukemia, and ized by the presence of a serum monoclonal
Waldenström’s macroglobulinemia. The clinical and immunoglobulin of the IgM class. Although anemia
laboratory data for these patients are summarized in was frequent (85%), they found cryoglobulinemia and
Tables 3-30 and 3-31. These authors used the term AIHA in only 5% of the patients, and stated that this
chronic CAS only when no abnormal cells were was too small a number of patients from which to draw
observed in the peripheral blood, bone marrow, or other meaningful conclusions.
examined tissues, and when no conversion to a malig-
nant condition occurred during the follow-up period. NONHEMATOLOGIC MALIGNANCIES
The specificity of the cold agglutinins varied from AND COLD AGGLUTININ SYNDROME
group to group. Whereas in lymphoma and
Waldenström’s macroglobulinemia, anti-I was The association of nonhematologic malignant dis-
unusual (32% to 34%), this specificity was present in orders with CAS is uncommon.213,427,429,434 Wortman
74% of patients with CAS. Indeed, Rosse772 stressed and coworkers434 reported seven patients who had a
that the finding of a cold agglutinin of unusual variety of nonhematologic malignancies including
specificity (e.g., anti-i) in a patient with seemingly squamous cell carcinoma of the lung, metastatic ade-
idiopathic CAS should alert one to the likely presence nocarcinoma of the adrenal, metastatic adenocarci-
of an underlying lymphoma. noma of the colon, a mixed parotid tumor, a carcinoid
Further findings of note are that Crisp and tumor of the ampulla of Vater, carcinoma of the
Pruzanski107 reported that in 71% of the patients with larynx, and a thoracic neurinoma. The high frequency
lymphoma, the type of light chain of cold agglutinins of carcinomas in patients in the age group in which
was λ, whereas in Waldenström’s macroglobulinemia CAS occurs suggests that the relationship may be for-
and chronic cold agglutinin disease, the majority of tuitous in at least some instances. That this is not true
cold agglutinins had κ light chains. Fifty-eight percent in all cases is indicated by the patient reported by
of IgM-κ cold agglutinins were anti-I, 25% were anti-i, Lands and Foust427 in whom resolution of the AIHA
and 17% had other specificities. Twenty-five percent occurred after excision of a renal cell carcinoma.
of IgM-λ cold agglutinins were anti-I, 50% were anti-i, The most effective management of AIHA in patients
and 25% had other specificities. with carcinomas is to treat the underlying malig-
Kyrtsonis and coworkers773 reviewed the clinical nancy.774 In cases in which tumor removal is not pos-
course and prognostic factors in 60 patients with sible, the AIHA should be treated independently as
Waldenström’s macroglobulinemia, which they described in Chapter 11.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 115

R E F E R E N C E S case report and review of the literature. Am J Hematol


1998;57:82−84.
1. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias. 25. Lipski DA, Bergamini TM: Upper extremity arterial thrombo-
New York: Churchill Livingstone, 1980. sis associated with immunohemolytic anemia. Surgery
2. Pirofsky B: Autoimmunization and the Autoimmune Hemol- 1996;119:354−355.
ytic Anemias. Baltimore: Williams & Wilkins Company, 1969. 26. David A, Thomas M: Upperextremity arterial thrombosis
3. Bottiger LE, Westerholm B: Acquired haemolytic anaemia. I: associated with immunohemolytic anemia. Surgery 1996;119:
Incidence and aetiology. Acta Med Scand 1973;193:223−226. 354−355.
4. Sokol RJ, Booker DJ, Stamps R: The pathology of autoimmune 27. Shiozawa Z, Ueda R, Mano T, Tsugane R, Kageyama N:
haemolytic anaemia. J Clin Pathol 1992;45:1047−1052. Superior sagittal sinus thrombosis associated with Evans’ syn-
5. Dacie JV, Worlledge SM: Auto-immune hemolytic anemias. In: drome of haemolytic anaemia. J Neurol 1985;232:280−282.
Brown EB, Moore CV (eds): Progress in Hematology. New 28. Kowal-Vern A, Radhakrishnan J, Goldman J, Hutchins W,
York: Grune & Stratton, 1969:82−120. Blank J: Mesenteric and portal vein thrombosis after splenec-
6. Engelfriet CP, Overbeeke MA, dem Borne AE: Autoimmune tomy for autoimmune hemolytic anemia. J Clin Gastroenterol
hemolytic anemia. Semin Hematol 1992;29:3−12. 1988;10:108−110.
7. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Warm- 29. Silberstein LE, Shoenfeld Y, Schwartz RS, Berkman EM: A
antibody syndromes, I: “Idiopathic” types: History and clini- combination of IgG and IgM autoantibodies in chronic cold
cal features. In: The Haemolytic Anaemias, 3rd ed. vol. 3, The agglutinin disease: Immunologic studies and response to sple-
Auto-Immune Haemolytic Anaemias. New York: Churchill nectomy. Vox Sang 1985;48:105–109.
Livingstone, 1992:6−53. 30. Freedman J, Lim FC, Musclow E, Fernandes B, Rother I:
8. Pirofsky B: Clinical aspects of autoimmune hemolytic anemia. Autoimmune hemolytic anemia with concurrence of warm
Semin Hematol 1976;13:251−265. and cold red cell autoantibodies and a warm hemolysin.
9. Allgood JW, Chaplin H, Jr: Idiopathic acquired autoimmune Transfusion 1985;25:368−372.
hemolytic anemia: A review of forty-seven cases treated from 31. Murayama A, Ayabe T, Toyama K, Nakamura Y: Pulmonary
1955 through 1965. Am J Med 1967;43:254−273. embolism and hemolytic anemia in a patient with cold agglu-
10. Dausset J, Colombani J: The serology and the prognosis of 128 tinin disease. Jpn Circ J 1981;45:1306−1308.
cases of autoimmune hemolytic anemia. Blood 1959;14:1280. 32. Saif MW, Morse EE, Greenberg BR: HIV-associated autoim-
11. Sokol RJ, Hewitt S, Stamps BK, Hitchen PA: Autoimmune mune hemolytic anemia complicated by pulmonary
haemolysis in childhood and adolescence. Acta Haematol embolism following a red blood cell transfusion: case report
1984;72:245−257. and review of the literature. Conn Med 1998;62:67−70.
12. Buchanan GR, Boxer LA, Nathan DG: The acute and transient 33. Bilgrami S, Cable R, Pisciotto P, Rowland F, Greenberg B: Fatal
nature of idiopathic immune hemolytic anemia in childhood. disseminated intravascular coagulation and pulmonary
J Pediatr 1976;88:780−783. thrombosis following blood transfusion in a patient with
13. Habibi B, Homberg JC, Schaison G, Salmon C: Autoimmune severe autoimmune hemolytic anemia and human immun-
hemolytic anemia in children: A review of 80 cases. Am J Med odeficiency virus infection. Transfusion 1994;34:248−252.
1974;56:61−69. 34. Feinstein DI: Inhibitors of blood coagulation. In: Hoffman R,
14. Berman LB: When the urine is red. JAMA 1977;237:2753−2754. Benz EJ, Shattil SJ, et al (eds): Hematology: Basic Principles and
15. Dameshek W, Schwartz SO: Acute hemolytic anemia Practice. Philadelphia: Churchill Livingstone, 2000:1963−1983.
(acquired hemolytic icterus, acute type). Medicine 35. Crosby WH, Rappaport H: Autoimmune hemolytic anemia.
1940;19:231. 1. Analysis of hematologic observations with particular refer-
16. Schwartz RS, Costea N: Autoimmune hemolytic anemia: ence to their prognostic value: A survey of 57 cases. Blood
Clinical correlations and biological implications. Semin 1957;12:42.
Hematol 1966;3:2−26. 36. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Warm-
17. Dacie JV: Auto-immune haemolytic anaemia (AIHA): antibody syndromes II: “idiopathic” types: haematological
Pathogenesis. In: The Haemolytic Anaemias, 3rd ed. vol. 3, and biochemical findings. In: The Haemolytic Anaemias, 3rd
The Auto-Immune Haemolytic Anaemias. New York: ed. vol. 3, The Auto-Immune Haemolytic Anaemias. New
Churchill Livingstone, 1992:392−451. York: Churchill Livingstone, 1992:54−93.
18. Shirey RS, Kickler TS, Bell W, Little B, Smith B, Ness PM: Fatal 37. Waterbury L, Parnes H, Katz RS: Fatal warm antibody
immune hemolytic anemia and hepatic failure associated with autoimmune hemolysis with marked erythrocyte autoaggluti-
a warm-reacting IgM autoantibody. Vox Sang 1987;52:219−222. nation and liver necrosis. South Med J 1986;79:646−647.
19. Swisher SN: Acquired hemolytic disease. Postgrad Med 38. Hargraves MM, Herrell WE, Pearman RO: Erythrophagocytic
1966;40:378−386. anemia (Lederer’s anemia?): Report of a case with recovery.
20. Crosby WH, Rappaport H: Autoimmune hemolytic anemia. Proc Staff Meetings Mayo Clin 1941;16:107−112.
Analysis of hematologic observations with particular refer- 39. Berry BH, Watson JD: Acquired hemolytic anemia: Report
ence to their prognostic value: A survey of 57 cases. Blood of a case in an infant with discussion of several etiological
1956;12:42−55. factors which may have been operative. Arch Pediatr
21. Pullarkat V, Ngo M, Iqbal S, Espina B, Liebman HA: Detection 1951;68:10−27.
of lupus anticoagulant identifies patients with autoimmune 40. Zinkham WH, Diamond LK: In vitro erythrophagocytosis in
haemolytic anaemia at increased risk for venous thromboem- acquired hemolytic anemia. Blood 1952;7:592−601.
bolism. Br J Haematol 2002;118:1166−1169. 41. de Gruchy GC: The diagnosis and management of acquired
22. Kokori SI, Ioannidis JP, Voulgarelis M, Tzioufas AG, haemolytic anaemia. Aust Ann Med 1954;3:106−115.
Moutsopoulos HM: Autoimmune hemolytic anemia in 42. Pisciotta AV, Downer EM, Hinz JE: Clinical and laboratory
patients with systemic lupus erythematosus. Am J Med correlation in severe autoimmune hemolytic anemia. Arch
2000;108:198−204. Intern Med 1959;104:264−276.
22a. Galli M, Luciani D, Bertolini G, Barbui T: Lupus anticoagu- 43. Greenberg J, Curtis-Cohen M, Gill FM, Cohen A: Prolonged
lants are stronger risk factors for thrombosis than anticar- reticulocytopenia in autoimmune hemolytic anemia of child-
diolipin antibodies in the antiphospholipid syndrome: A hood. J Pediatr 1980;97:784−786.
systematic review of the literature. Blood 2003;101:1827–1832. 44. Liesveld JL, Rowe JM, Lichtman MA: Variability of the ery-
23. Borgna PC, Carnelli V, Caruso V, et al: Thromboembolic events thropoietic response in autoimmune hemolytic anemia:
in beta thalassemia major: An Italian multicenter study. Acta Analysis of 109 cases. Blood 1987;69:820−826.
Haematol 1998;99:76−79. 45. Heisel MA, Ortega JA: Factors influencing prognosis in child-
24. Hayag-Barin JE, Smith RE, Tucker FC, Jr: Hereditary sphero- hood autoimmune hemolytic anemia. Am J Pediatr Hematol
cytosis, thrombocytosis, and chronic pulmonary emboli: A Oncol 1983;5:147−152.
116 Immune Hemolytic Anemias

46. Evans RS, Duane RT: Acquired hemolytic anemia. I: The 71. Lees MH, Fisher OD, Sharks JM: Case of immune aplastic
relation of erythrocyte antibody to activity of the disease. haemolytic anaemia with thrombocytopenia. Brit Med J
II: Significance of thrombocytopenia and leukopenia. Blood 1960;1:110.
1949;4:1196−1213. 72. Conley CL, Lippman SM, Ness P: Autoimmune hemolytic
47. Ciaffoni S, Ferro I, Potenza R, Campo G: Evans syndrome: A anemia with reticulocytopenia: A medical emergency. JAMA
case of autoimmune thrombocytopenia and autoimmune 1980;244:1688−1690.
hemolytic anemia caused by anti-Jka. Haematologica 73. Hegde UM, Gordon-Smith EC, Worlledge SM: Reticu-
1987;72:245−247. locytopenia and “absence” of red cell autoantibodies in
48. Monch H, Breithaupt H, Mueller-Eckhardt C: Immunological immune haemolytic anaemia. Br Med J 1977;2:1444−1447.
studies in a case of Evans’ syndrome. Blut 1981;42:27−32. 74. Gundersen SG, Bjoerneklett A, Bruun JN: Severe erythro-
49. Wang WC: Evans syndrome in childhood: Pathophysiology, blastopenia and hemolytic anemia during a hepatitis A infec-
clinical course, and treatment. Am J Pediatr Hematol Oncol tion. Scand J Infect Dis 1989;21:225−228.
1988;10:330−338. 75. Crosby WH, Rappaport H: Reticulocytopenia in autoimmune
50. Ng SC: Evans syndrome: A report on 12 patients. Clin Lab hemolytic anemia. Blood 1956;11:929−936.
Haematol 1992;14:189−193. 76. Celada A: Autoimmune haemolytic anaemia and aplastic
51. Mathew P, Chen G, Wang W: Evans syndrome: Results of a crisis. Br J Haematol 1984;57:178−190.
national survey. J Pediatr Hematol Oncol 1997;19:433−437. 77. Harley J, Dods L: Some acquired haemolytic anaemias of
52. Savasan S, Warrier I, Ravindranath Y: The spectrum of Evans’ childnood. Annals of Medicine 1959;8:98−108.
syndrome. Arch Dis Child 1997;77:245−248. 78. Meyer RJ, Hoffman R, Zanjani ED: Autoimmune hemolytic
53. Evans RS, Takahashi K, Duane RT, Payne R, Liu C-K: Primary anemia and periodic pure red cell aplasia in systemic lupus
thrombocytopenic purpura and acquired hemolytic anemia: erythematosus. Am J Med 1978;65:342−345.
Evidence for a common etiology. Arch Intern Med 1951; 79. Lees MH, Fisher OD, Shanks JM: Case of immune aplastic
87:48−65. haemolytic anaemia with thrombocytopenia. Br Med J
54. Zucker-Franklin D, Karpatkin S: Red-cell and platelet frag- 1960;1:110−111.
mentation in idiopathic autoimmune thrombocytopenic 80. Tomiyama J, Adachi Y, Hanada T, Matsunaga Y: Human par-
purpura. N Engl J Med 1977;297:517−523. vovirus B19-induced aplastic crisis in autoimmune haemolytic
55. Waugh TR: Acquired haemolytic jaundice in a woman previ- anaemia. Br J Haematol 1988;69:288−289.
ously splenectomized for essential lthrombocytopenia. Folia 81. Lefrere JJ, Courouce AM, Bertrand Y, Girot R, Soulier JP:
haemat (Lpz) 1932;48:248−260. Human parvovirus and aplastic crisis in chronic hemolytic
56. Pui CH, Wilimas J, Wang W: Evans syndrome in childhood. anemias: A study of 24 observations. Am J Hematol
J Pediatr 1980;97:754−758. 1986;23:271−275.
57. Crosby WH: The clinical aspects of immunologic hemolytic 82. Bertrand Y, Lefrere JJ, Leverger G, et al: Autoimmune
anemia. Vox Sang 1955;26:3. haemolytic anaemia revealed by human parvovirus linked
58. Chertkow G, Dacie JV: Results of splenectomy in auto-immune erythroblastopenia. Lancet 1985;ii:382−383.
haemolytic anaemia. Br J Haematol 1956;2:237. 83. Pisciotta AV, Hinz JE: Occurrence of agglutinogens in nor-
59. Fagiolo E: Platelet and leukocyte antibodies in autoimmune moblasts. Proc Soc Exp Biol Med 1956;91:356.
hemolytic anemia. Acta Haematol 1976;56:97−106. 84. Yunis JJ, Yunis E: Cell antigens and cell specialization. I: A
60. Pegels JG, Helmerhorst FM, van Leeuwen EF, van de Plas-van study of blood group antigens on normoblasts. Blood
Dalen, Engelfriet CP, dem Borne AE: The Evans syndrome: 1963;22:53.
Characterization of the responsible autoantibodies. Br J 85. Mangan KF, Besa EC, Shadduck RK, Tedrow H, Ray PK:
Haematol 1982;51:445−450. Demonstration of two distinct antibodies in autoimmune
61. Miller BA, Schultz Beardsley D: Autoimmune pancytopenia of hemolytic anemia with reticulocytopenia and red cell aplasia.
childhood associated with multisystem disease manifesta- Exp Hematol 1984;12:788−793.
tions. J Pediatr 1983;103:877−881. 86. Brown KE: Haematological consequences of parvovirus
62. Chapman JF, Metcalfe P, Murphy MF, Burman JF, Waters AH: B19 infection. Baillieres Best Pract Res Clin Haematol
Sequential development of platelet, neutrophil and red cell 2000;13:245−259.
autoantibodies associated with measles infection. Clin Lab 87. Elsacker-Niele AM, Kroes AC: Human parvovirus B19:
Haematol 1984;6:219−228. Relevance in internal medicine. Neth J Med 1999;54:221−230.
63. Wiesneth M, Pflieger H, Frickhofen N, Heimpel H: Idiopathic 88. Pattison JR, Jones SE, Hodgson J, et al: Parvovirus infections
combined immunocytopenia. Br J Haematol 1985;61:339−348. and hypoplastic crisis in sickle-cell anaemia. Lancet
64. Conley CL, Lippman SM, Ness PM, Petz LD, Branch DR, 1981;1:664−665.
Gallagher MT: Autoimmune hemolytic anemia with reticulo- 89. Brown KE, Hibbs JR, Gallinella G, et al: Resistance to
cytopenia and erythroid marrow. N Engl J Med 1982;306: parvovirus B19 infection due to lack of virus receptor
281−286. (erythrocyte P antigen). N Engl J Med 1994;330:1192−1196.
65. Greenberg J, Curtis-Cohen M, Gill FM, Cohen A: Prolonged 90. Heegaard ED, Brown KE: Human parvovirus B19. Clin
reticulocytopenia in autoimmune hemolytic anemia of child- Microbiol Rev 2002;15:485−505.
hood. J Pediatr 1980;97:784−786. 91. Young N: Hematologic and hematopoietic consequences of
66. Hauke G, Fauser AA, Weber S, Maas D: Reticulocytopenia in B19 parvovirus infection. Semin Hematol 1988;25:159−172.
severe autoimmune hemolytic anemia (AIHA) of the warm 92. Harris JW: Parvovirus B19 for the hematologist. Am J Hematol
antibody type. Blut 1983;46:321−327. 1992;39:119−130.
67. Olcay L, Duzova A, Gumruk F: A warm antibody mediated 93. Silverstein MN, Gomes MR, Elveback LR, ReMine WH,
acute hemolytic anemia with reticulocytopenia in a four- Linman JW: Idiopathic acquired hemolytic anemia: Survival in
month-old girl requiring immunosuppressive therapy. Turk J 117 cases. Arch Intern Med 1972;129:85−87.
Pediatr 1999;41:239−244. 94. Dacie JV: Auto-immune haemolytic anaemias (AIHA): treat-
68. Carapella dL, Stegagno M, Morino G, Ballati G, Lorenzini F: ment. In The Haemolytic Anaemias, 3rd ed. vol. 3, The Auto-
Prolonged reticulocytopenia and transient erythroid hypopla- Immune Haemolytic Anaemias. New York: Churchill
sia in a child with autoimmune haemolytic anaemia. Livingstone, 1992:452−520.
Haematologica 1984;69:315−320. 95. Crosby WH, Rappaport H: Autoimmune hemolytic anemia. 1.
69. Burston J, Husain OA, Hutt MS, Tanner EI: Two cases of Analysis of hematologic observations with particular reference
autoimmune haemolysis and aplasia. Brit Med J 1959;1:83. to their prognostic value; a survey of 57 cases. Blood 1957;
70. Eisemann G, Dameshek W: Splenectomy for pure red-cell 12:42.
hypoplastic and aregenerative anemia associated with autoim- 96. Worlledge S: Immune haemolytic anemias. In Hardisty RM,
mune hemolytic disease;report of a case. New Engl J Med Weatherall DJ (eds): Blood and Its Disorders. Oxford:
1954;251:1044. Blackwell, 1974:714.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 117

97. Poschmann A, Fischer K: Autoimmune haemolytic anaemia: tional disorders associated with intravascular coagulation
Recent advances in pathogenesis, diagnosis and treatment. necrosis of the skin. J Am Acad Dermatol 1992;272 Pt 1.:277.
Eur J Pediatr 1985;143:258−260. 123. Barth JH: Infectious mononucleosis (glandular fever) compli-
98. Heisel MA, Ortega JA: Factors influencing prognosis in child- cated by cold agglutinins, cold urticaria and leg ulceration.
hood autoimmune hemolytic anemia. Am J Pediatr Hematol Acta Derm Venereol 1981;61:451−452.
Oncol 1983;5:147−152. 124. Packman CH: Cryopathic hemolytic syndromes. In Beutler E,
99. Miyazaki S, Nakayama K, Akabane T, et al: Follow-up study of Lichtman MA, Coller BS, Kipps TJ (eds): Williams
34 children with autoimmune hemolytic anemia. Nippon Hematology. New York: McGraw-Hill, 1995:649−655.
Ketsueki Gakkai Zasshi 1983;46:6−10. 125. Thomas TD, Donofrio PD, Angelo J: Peripheral neuropathy in
100. Van Loghem JJ, Van der Hart M, Dorfmeier H: Serological cold agglutinin disease. Muscle Nerve 1991;14:331−334.
studies in acquired hemolytic anemia. Proceedings of the Sixth 126. Hattersley PG, Gerard PW, Caggiano V, Nash DR: Erroneous
International Congress of the International Society of values on the Model S Coulter Counter due to high titer cold
Hematology. New York: Grune & Stratton, 1958. autoagglutinins. Am J Clin Pathol 1971;55:442−446.
101. Worlledge S: Auto-immunity and blood diseases. Practitioner 127. Petrucci JV, Dunne PA, Chapman CC: Spurious erythrocyte
1967;199:171−179. indices as measured by the model S Coulter counter due to
102. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Cold- cold agglutinins. Am J Clin Pathol 1971;56:500−502.
antibody syndromes. I: Idiopathic types: Clinical presentation 128. DeLange JA, Eernisse GJ, Veltkamp JJ: Cold agglutinins
and haematological and serological findings. In The Haemolytic and the Coulter Counter Model S. Am J Clin Pathol 1972;
Anaemias, 3rd ed. vol. 3, The Auto-Immune Haemolytic 58:599−600.
Anaemias. New York: Churchill Livingstone, 1992:210−239. 129. Lawrence C, Zozicky O: Spurious red-cell values with the
103. Schubothe H: The cold hemagglutinin disease. Semin Hematol Coulter Counter. N Engl J Med 1983;309:925−926.
1966;3:27−47. 130. Roelcke D: Cold agglutination. Transfus Med Rev 1989;3:
104. Zupanska B, Lawkowicz W, Gorska B, et al: Autoimmune 140−166.
haemolytic anaemia in children. Br J Haematol 1976;34:511− 131. Leddy JP, Swisher SN: Acquired immune hemolytic disorders
520. (including drug-induced immune hemolytic anemia). In:
105. McNicholl FP: Clinical syndromes associated with cold agglu- Samter M (ed): Immunologic Diseases. Boston: Little, Brown &
tinins. Transfus Sci 2000;22:125−133. Co., 1978:1187.
106. Nydegger UE, Kazatchkine MD, Miescher PA: Immuno- 132. Schreiber AD, Herskovitz BS, Goldwein M: Low-titer cold-
pathologic and clinical features of hemolytic anemia due to hemagglutinin disease: Mechanism of hemolysis and response
cold agglutinins. Seminars Hematol 1991;28:66−77. to corticosteroids. N Engl J Med 1977;296:1490−1494.
107. Crisp D, Pruzanski W: B-cell neoplasms with homogeneous 133. Rosse WF: Correlation of in vivo and in vitro measurements of
cold-reacting antibodies (cold agglutinins). Am J Med 1982; hemolysis in hemolytic anemia due to immune reactions. Prog
72:915−922. Hematol 1973;8:51−75.
108. Niejadlik DC, Lozner EL: Cooling mattress induced acute 134. Chitnavis VN, Patou G, Makar YF, Kendra JR: B19 parvovirus
hemolytic anemia. Transfusion 1974;14:145−147. induced red cell aplasia complicating acute cold antibody
109. Colmers RA, Snavely JG: Acute hemolytic anemia in primary mediated haemolytic anaemia. Br J Haematol 1990;76:433−434.
atypical pneumonia produced by exposure and chilling. 135. Cazzola M, Barosi G, Ascari E: Cold haemagglutinin disease
N Engl J Med 1947;237:505−510. with severe anaemia, reticulocytopenia and erythroid bone
110. Ferriman DG, Dacie JV, Keele KD, Fullerton JM: The associa- marrow. Scand J Haematol 1983;30:25−29.
tion of Raynaud’s phenomena, chronic haemolytic anaemia, 136. Samson D, Halliday D, Nicholson DC, Chanarin I: Quantitation
and the formation of cold antibodies. Quart J Med 1951; of ineffective erythropoiesis from the incorporation of [15N]
20:275. delta-aminolaevulinic acid and [15N] glycin into early labelled
111. Lauchli S, Widmer L, Lautenschlager S: Cold agglutinin bilirubin. II: Anaemic patients. Br J Haematol 1976;34:45−53.
disease—the importance of cutaneous signs. Dermatology 137. Fudenberg HH, Kunkel HG: Physical properties of the red cell
2001;202:356−358. agglutinins in acquired hemolytic anaemia. J Exp Med
112. Barcroft H, Edholm OG: Temperature and blood flow in the 1957;1:689.
human forearm. J Physiol (Lond) 1946;104:366. 138. Schwartz T, Jager BV: Cryoglobulinemia and Raynaud’s syn-
113. Harboe M: Cold auto-agglutinins. Vox Sang 1971;20:289−305. drome in a case of chronic lymphocytic leukemia. Cancer
114. Mitchell AB, Pergrum GD, Gill AM: Cold agglutinin disease 1949;2:319.
with Raynaud’s phenomenon. Proc R Soc Med 1974;67:113−115. 139. Ulvestad E, Berentsen S, Bo K, Shammas FV: Clinical
115. Poldre P, Pruzanski W, Chiu HM, Dotten DA: Fulminant gan- immunology of chronic cold agglutinin disease. Eur J
grene in transient cold agglutinemia associated with Haematol 1999;63:259−266.
Escherichia coli infection. Can Med Assoc J 1985;132:261−263. 140. Evans RS, Baxter E, Gilliland BC: Chronic hemolytic anemia
116. Pruzanski W, Shumak KH: Biologic activity of cold-reacting due to cold agglutinins: A 20-year history of benign gammopa-
autoantibodies (first of two parts). N Engl J Med 1977;297: thy with response to chlorambucil. Blood 1973;42:463−470.
538−542. 141. Seldon M, Isbister JP, Raik E, Biggs JC: A fatal case of cold auto-
117. Pruzanski W, Shumak KH: Biologic activity of cold-reacting immune hemolytic anemia. Am J Clin Pathol 1980;73: 716−717.
autoantibodies (second of two parts). N Engl J Med 142. Rousey SR, Smith RE: A fatal case of low titer anti-PR cold
1977;297:583−589. agglutinin disease. Am J Hematol 1990;35:286−287.
118. Pruzanski W, Cowan DH, Parr DM: Clinical and immuno- 143. Mandigers CM, Keuning JJ, Booij AC: A patient with fatal cold
chemical studies of IgM cold agglutinins with lambda type haemagglutinins. Neth J Med 1996;49:209−211.
light chains. Clin Immunol Immunopathol 1974;2:234−245. 144. Howard CP, Mills ES: Paroxysmal hemoglobinuria. Amer J
119. Rorvik K: The syndrome of high-titre cold haemag- Med Sci 1938;196:792.
glutination: A survey and a case report. Acta Med Scand 145. Becker RM: Paroxysmal cold hemoglobinurias. Arch Intern
1954;148:299. Med 1948;81:630.
120. Gaddy CG, Powell LW: Raynaud’s syndrome associated with 146. Johnsen HE, Brostrphom K, Madsen M: Paroxysmal cold
idiopathic cryoglobulinemia and cold agglutinins: Report of a haemoglobinuria in children: 3 cases encountered within a
case and discussion of classification of cyroglobulinemia. Arch period of 7 months. Scand J Haematol 1978;20:413–416.
Intern Med 1958;102:468. 147. Bird GW, Wingham J, Martin AJ, et al: Idiopathic non-
121. Stone MS, Piette WW, Davey WP: Cutaneous necrosis at sites syphilitic paroxysmal cold haemoglobinuria in children. J Clin
of transfusion: Cold agglutinin disease. J Am Acad Dermatol Pathol 1976;29:215−218.
1988;192 Pt 1.:356−357. 148. Gottsche B, Salama A, Mueller-Eckhardt C: Donath-
122. Zouboulis CC, Gollnick H, Orfanos CE: Essential thrombo- Landsteiner autoimmune hemolytic anemia in children. A
cythemia and idiopathic cold agglutinin disease: Two addi- study of 22 cases. Vox Sang 1990;58:281−286.
118 Immune Hemolytic Anemias

149. Wynn RF, Stevens RF, Bolton-Maggs PH, Schwe K, Will AM: 176. Wishart MM, Davey MG: Infectious mononucleosis
Paroxysmal cold haemoglobinuria of childhood: A review of complicated by acute haemolytic anaemia with a
the management and unsual presenting features of six cases. positive Donath-Landsteiner reaction. J Clin Pathol
Clin Lab Haematol 1998;20:373–375. 1973;26:332−334.
150. Lambert T, Noblins M, Tchernia G: Biphasic hemolysin 177. Burkhart PT, Hsu TCS: IgM cold-warm hemolysins in infec-
hemolytic anemia in children. Am J Hematol 1993;43:77. tious mononucleosis. Transfusion 1979;19:535−538.
151. Nordhagen R, Stensvold K, Winsnes A, Skyberg D, Storen A: 178. Uzokwe CO, Gwynn AM, Gorst DW, Adamson AR: Infectious
Paroxysmal cold haemoglobinuria: The most frequent acute mononucleosis complicated by haemolytic anaemia due to the
autoimmune haemolytic anaemia in children? Acta Paediatr Donath Landsteiner antibody and by severe neutropenia. Clin
Scand 1984;73:258−262. Lab Haematol 1993;15:137−140.
152. Heddle NM: Acute paroxysmal cold hemoglobinuria. Transfus 179. Engelfriet CP, Beckers D, Borne AE, Reynierse E, Van Loghem
Med Rev 1989;3:219−229. JJ: Haemolysins probably recognizing the antigen p. Vox Sang
153. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Cold- 1972;23:176−181.
antibody syndromes V: Paroxysmal cold haemoglobinuria 180. Boccardi V, D’Annibali S, Di Natale G, Girelli G, Summonti D:
(PCH). In The Haemolytic Anaemias, 3rd ed. vol. 3, The Mycoplasma pneumoniae infection complicated by paroxys-
Auto-Immune Haemolytic Anaemias. New York: Churchill mal cold hemoglobinuria with anti-P specificity of biphasic
Livingstone, 1992:329−362. hemolysin. Blut 1977;34:211−214.
154. Sokol RJ, Hewitt S, Stamps BK: Autoimmune haemolysis asso- 181. Lau P, Sererat S, Moore V, McLeish K, Alousi M: Paroxysmal
ciated with Donath-Landsteiner antibodies. Acta Haematol cold hemoglobinuria in a patient with Klebsiella pneumonia.
1982;68:268−277. Vox Sang 1983;44:167−172.
155. Sokol RJ, Booker DJ, Stamps R: Erythropoiesis: Paroxysmal cold 182. Brown KE, Anderson SM, Young NS: Erythrocyte P antigen:
haemoglobinuria: A clinico-pathological study of patients with Cellular receptor for B19 parvovirus. Science 1993;262:
a positive Donath-Landsteiner test. Hematol 1999;4:137−164. 114−117.
156. Donath J, Landsteiner K: Uber Kaltehaemoglobinurie. Ergebn 183. Sharpe JS, Booker DJ, Stamps R, Sokol RJ: Parvovirus B19
Hyg Bakt 1925;7:184. infection and paroxysmal cold haemoglobinuria. Transfus
157. Nabarro D: Congenital Syphilis. London: Arnold, 1954. Med 1996;6(Suppl 2):24 (Abstract).
158. Donath J, Landsteiner K: Weitere Beobactungen uber paroxys- 184. Lindgren S, Zimmerman S, Gibbs F, Garratty G: An unusual
male Haemoglobinurie. Abl Bakt (Abt I Orig) 1908;45:204. Donath-Landsteiner antibody detectable at 37°C by the
159. Sharara AI, Hillsley RE, Wax TD, Rosse WF: Paroxysmal cold antiglobulin test. Transfusion 1985;25:142−144.
hemoglobinuria associated with non-Hodgkin’s lymphoma. 185. Johnsen HE, Brostrphom K, Madsen M: Paroxysmal cold
South Med J 1994;87:397−399. haemoglobinuria in children: 3 cases encountered within a
160. Sivakumaran M, Murphy PT, Booker DJ, Wood JK, Stamps R, period of 7 months. Scand J Haematol 1978;20:413−416.
Sokol RJ: Paroxysmal cold haemoglobinuria caused by non- 186. Lindgren S, Zimmerman S, Gibbs F, Garratty G: An unusual
Hodgkin’s lymphoma. Br J Haematol 1999;105:278−279. Donath-Landsteiner antibody detectable at 37 degrees C by
161. Lippman SM, Winn L, Grumet FC, Levitt LJ: Evans’ syndrome the antiglobulin test. Transfusion 1985;25:142−144.
as a presenting manifestation of atypical paroxysmal cold 187. Vreman HJ, Mahoney JJ, Stevenson DK: Carbon monoxide and
hemoglobinuria. Am J Med 1987;82:1065−1072. carboxyhemoglobin. Adv Pediatr 1995;42:303−334.
162. Patel M, Durao H, Govender Y: Paroxysmal cold haemoglo- 188. Wynn RF, Stevens RF, Bolton-Maggs PH, Schwe K, Will AM:
binuria coexisting with cold agglutinins in a patient with Paroxysmal cold haemoglobinuria of childhood: A review of
syphilis resulting in peripheral gangrene: A case report. East the management and unusual presenting features of six cases.
Afr Med J 1993;70:526−527. Clin Lab Haematol 1998;20:373−375.
163. Banov CH: Paroxysmal cold hemoglobinuria: Apparent remis- 189. Bell CA, Zwicker H, Sacks HJ: Autoimmune hemolytic
sion after splenectomy. JAMA 1960;174:1974. anemia: Routine serologic evaluation in a general hospital
164. Carswell J, Eloff P, Edkins P, Willmore A: Paroxysmal cold population. Am J Clin Pathol 1973;60:903−911.
haemoglobinuria: A case report. S Afr Med J 1979;55:891−892. 190. Garratty G: Erythrophagocytosis on the peripheral blood
165. Nelson MG, Nicholl B: Paroxysmal cold haemoglobinuria. smear and paroxysmal cold hemoglobinuria. Transfusion
Irish J Med Sci 1960; 6th series:49−57. 2001;41:1073−1074.
166. Shah AA, Desai AB: Paroxysmal cold hemoglobinuria (case 191. Hernandez JA, Steane SM: Erythrophagocytosis by segmented
report). Indian Pediatr 1977;14:219−221. neutrophils in paroxysmal cold hemoglobinuria. Am J Clin
167. Wood JK: Paroxysmal cold haemoglobinuria in Jamaica: A case Pathol 1984;81:787−789.
report. West Indian Med J 1968;17:175−179. 192. Gelfand EW, Abramson N, Segel GB, Nathan DG: Buffy-coat
168. Kumar ND, Sethi S, Pandhi RK: Paroxysmal cold haemoglo- observations and red-cell antibodies in acquired hemolytic
binuria in syphilis patients. Genitourin Med 1993;69:76. anemia. N Engl J Med 1971;284:1250−1252.
169. Bunch C, Schwartz FC, Bird GW: Paroxysmal cold haemoglo- 193. Friedman HD, Dracker RA: Intravascular erythrophagocyto-
binuria following measles immunization. Arch Dis Child sis. JAMA 1991;265:1082.
1972;47:299−300. 194. Garratty G, Nance S, Arndt P, et al: Positive direct monocyte
170. O’Neill BJ, Marshall WC: Paroxysmal cold haemoglobinuria monolayer assays associated with positive Donath
and measles. Arch Dis Child 1967;42:183−186. Landsteiner tests. Transfusion 1989;29(Suppl):49S. (Abstract)
171. Colley EW: Paroxysmal cold haemoglobinuria after mumps. 195. Harrison SJ, Simpson E: Images in haematology. Erythro-
Br Med J 1964;1:1552−1553. phagocytosis. Br J Haematol 2001;113:2.
172. Papalia MA, Schwarer AP: Paroxysmal cold haemoglobinuria 196. Lobreglio G, Valacca A: Images in clinical medicine.
in an adult with chicken pox. Br J Haematol 2000;109: Erythrophagocytosis. N Engl J Med 2001;344:897.
328−329. 197. Depcik-Smith ND, Escobar MA, Ma AD, Brecher ME: RBC
173. Nakamura H, Watanabe T, Hayashida T, Ichimaru M: Donath- rosetting and erythrophagocytosis in adult paroxysmal cold
Landsteiner antibody of the IgM class with anti-I specificity hemoglobinuria. Transfusion 2001;41:163.
and possible efficacy of azathioprine therapy in paroxysmal 198. Uchida H: Uber die Erythrophagozytose der Leukozyten,
cold hemoglobinuria: A case report. Rinsho Ketsueki besonders bei der paroxysmalen Haemoglobinurie. Mitt med
1990;31:1548−1552. Fak 1921;26:503.
174. Chambers LA, Rauck AM: Acute transient hemolytic anemia 199. Montagnani M: Crise hemoclassique et hemoglobinurie parox-
with a positive Donath-Landsteiner test following parvovirus ystique. Presse Med 1921;29:1017.
B19 infection. J Pediatr Hematol Oncol 1996;18:178−181. 200. Ries CA, Garratty G, Petz LD, Fudenberg HH: Paroxysmal
175. Wolach B, Heddle N, Barr RD, Zipursky A, Pai KR, Blajchman cold hemoglobinuria: Report of a case with an exceptionally
MA: Transient Donath-Landsteiner haemolytic anaemia. Br J high thermal range Donath-Landsteiner antibody. Blood
Haematol 1981;48:425−434. 1971;38:491−499.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 119

201. Schmidt J, Clifford GO: Studies of the antibody in paroxysmal patient with chronic lymphocytic leukemia/small cell lym-
cold hemoglobinuria. Clinical Research 8, 217. 1960. (Abstract) phoma. Vox Sang 1998;74:122−126.
202. Kissmeyer-Nielson E, Schleisner P: Paroxysmal cold haemo- 225. Patten E, Reuter FP: Evans’ syndrome: Possible benefit from
globinuria of non-syphilitic type. Report of a case with plasma exchange. Transfusion 1980;20:589−593.
haemolytic anaemia and a normal cold haemagglutinin titre. 226. Sokol RJ, Hewitt S, Stamps BK: Autoimmune haemolysis: An
Dan med Bull 1963;10:52−58. 18-year study of 865 cases referred to a regional transfusion
203. Sirchia G, Ferrone S, Mercuriali F: A case of paroxysmal cold centre. Br Med J (Clin Res Ed) 1981;282:2023−2027.
haemoglobinuria with unusual serological findings. Haematol 227. Morselli M, Luppi M, Potenza L, et al: Mixed warm and cold
Lat 1965;8:103−112. autoimmune hemolytic anemia: Complete recovery after 2
204. Fisher S: Paroxysmal cold hemoglobinuria. Case report with courses of rituximab treatment. Blood 2002;99:3478−3479.
characterization of the Donath-Landsteiner hemolysin. Am J 228. Barry KG, Crosby WH: Auto-immune hemolytic anemia
Med 1969;46:475−479. arrested by removal of an ovarian teratoma: Review of the liter-
205. Djaldetti M, Elion D, Bessler H, Fishman P: Paroxysmal cold ature and report of a case. Ann Intern Med 1957;47:1002−1007.
hemoglobinuria. Transmission and scanning electron 229. Dawson MA, Talbert W, Yarbro JW: Hemolytic anemia associ-
microscopy features of erythrocytes. Am J Clin Pathol ated with an ovarian tumor. Am J Med 1971;50:552−556.
1975;63:804−810. 230. de Bruyere M, Sokal G, Devoitille JM, Fauchet-Dutrieux MC,
206. Rosse WF: Autoimmune hemolytic anemia due to cold-reacting De S, V: Autoimmune haemolytic anaemia and ovarian
antibodies: Paroxysmal cold hemoglobinuria. Clinical Immuno- tumour. Br J Haematol 1971;20:83−94.
hematology: Basic Concepts and Clinical Applications. Boston: 231. Payne D, Muss HB, Homesley HD, Jobson VW, Baird FG:
Blackwell Scientific Publications, 1990:583−589. Autoimmune hemolytic anemia and ovarian dermoid cysts:
207. Dacie JV: The Haemolytic Anaemias. Part II: Auto-Immune Case report and review of the literature. Cancer 1981;48:721−724.
Haemolytic Anaemias, 2nd ed. New York: Grune & Stratton, 232. Estrada CA, Lyons S, Terebelo H: Autoimmune hemolytic
1962. anemia and rheumatoid arthritis. South Med J 1990;83:599−600.
208. Sokol RJ, Hewitt S: Autoimmune hemolysis: A critical review. 233. Shashaty GG, Rath CE, Britt EJ: Autoimmune hemolytic
Crit Rev Oncol Hematol 1985;4:125−154. anemia associated with ulcerative colitis. Am J Hematol
209. Sokol RJ, Hewitt S, Booker DJ: Erythrocyte autoantibodies, 1977;3:199−208.
autoimmune haemolysis, and myelodysplastic syndromes. 234. Veloso FT, Fraga J, Carvalho J, Dias LM, Salgado MC:
J Clin Pathol 1989;42:1088−1091. Autoimmune hemolytic anemia in ulcerative colitis. A case
210. Hsu HC, Lin CK, Chau WK, Hu HY, Liu SM: Combined cold- report with review of the literature. J Clin Gastroenterol
and warm-antibody autoimmune hemolytic anemia⎯review 1991;13:445−447.
of the literature and a case report. Gaoxiong Yi Xue Ke Xue Za 235. Ramakrishna R, Manoharan A: Auto-immune haemolytic
Zhi 1989;5:350−356. anaemia in ulcerative colitis. Acta Haematol 1994;91:99−102.
211. Sheth SS, Karande SC, Nadkarni VB, Lahiri K, Jain MK, Shah 236. Blajchman MA, Hobbs JR, Dacie JV, Pettit JE, Worlledge SM:
MD: Autoimmune hemolytic anemia⎯mixed type. Indian Immunoglobulins in warm-type autoimmune haemolytic
Pediatr 1991;28:303−304. anaemia. Lancet 1969;2:340−344.
212. Zupanska B, Sokol RJ, Booker DJ, Stamps R: Erythrocyte 237. Dacie JV: Autoimmune hemolytic anemia. Arch Intern Med
autoantibodies, the monocyte monolayer assay and in vivo 1975;135:1293−1300.
haemolysis. Br J Haematol 1993;84:144−150. 238. Fudenberg HH: Immunologic deficiency, autoimmune
213. Sokol RJ, Booker DJ, Stamps R: Erythrocyte autoantibodies, disease, and lymphoma: Observations, implications, and spec-
autoimmune haemolysis, and carcinoma. J Clin Pathol ulations. Arthritis Rheum 1966;9:464−472.
1994;47:340−343. 239. Fudenberg HH: Genetically determined immune deficiency as
214. Crookston JH: Hemolytic anemia with IgG and IgM autoanti- the predisposing cause of “autoimmunity” and lymphoid neo-
bodies and alloantibodies. Arch Intern Med 1975;135:1314. plasia. Am J Med 1971;51:295−298.
215. Moake JL, Schultz DR: Hemolytic anemia associated with 240. Fudenberg HH: Are autoimmune diseases and malignancy
multiple autoantibodies and low serum complement. Am J due to selective T-cell deficiencies? Critical Factors in Cancer
Med 1975;58:431−437. Immunology. New York: Academic Press, 1975.
216. Sokol RJ, Hewitt S, Stamps BK: Autoimmune hemolysis: 241. Kruger J, Rahman A, Mogk KU, Mueller-Eckhardt C: T cell
Mixed warm and cold antibody type. Acta Haematologica deficiency in patients with autoimmune hemolytic anemia
1983;69:266−274. (“warm type”). Vox Sang 1976;31:1−12.
217. Shulman IA, Branch DR, Nelson JM, Thompson JC, Saxena S, 242. Rennert OM: The hypogammaglobulinemias. Ann Clin Lab Sci
Petz LD: Autoimmune hemolytic anemia with both cold and 1978;8:276−282.
warm autoantibodies. JAMA 1985;253:1746−1748. 243. Dacie JV: The Haemolytic Anaemias, 3rd ed. vol. 4, Secondary
218. Nusbaum NJ, Khosla S: Autoimmune hemolytic anemia with or Symptomatic Haemolytic Anaemias. New York: Churchill
both cold and warm autoantibodies. JAMA 1985;254:1175−1176. Livingstone, 1995.
219. Kajii E, Miura Y, Ikemoto S: Characterization of autoantibodies 244. Dameshek W, Komninos ZD: The present status of treatment
in mixed-type autoimmune hemolytic anemia. Vox Sang of autoimmune hemolytic anemia with ACTH and cortisone.
1991;60:45−52. Blood 1956;11:648.
220. De Angelis V, Biasinutto C, Pradella P, Errante D: Mixed-type 245. Evans RS, Weiser RS: The serology of autoimmune hemolytic
auto-immune haemolytic anaemia in a patient with HIV infec- disesase: Observations on forty-one patients. Arch Intern Med
tion. Vox Sang 1995;68:191−194. 1957;100:371.
221. Koduri PR, Singa P, Nikolinakos P: Autoimmune hemolytic 246. Chaplin H, Avioli LV: Grand rounds: Autoimmune hemolytic
anemia in patients infected with human immunodeficiency anemia. Arch Int Med 1977;137:346−351.
virus−1. Am J Hematol 2002;70:174−176. 247. West-Watson WN, Young CJ: Failed splenectomy in acholuric
222. Chen CY, Lu CL, Chiu CF, Chang FY, Lee SD: Primary biliary jaundice, and the relation ot toxaemia to the haemolytic crisis.
cirrhosis associated with mixed type autoimmune hemolytic Br Med J 1938;1:1305.
anemia and sicca syndrome: A case report and review of liter- 248. Burkert L, Becker G, Pisciotta AV: Ovarian malignancy and
ature. Am J Gastroenterol 1997;92:1547−1549. hemolytic anemia: Demonstration of a hemolytic serum factor.
223. Gologan R, Dima I, Butoianu E, Nicoara S, Colita D: Ann Intern Med 1970;73:91−93.
Autoimmune hemolytic anemia with warm antibodies com- 249. Cobo F, Pereira A, Nomdedeu B, et al: Ovarian dermoid cyst-
plicated with an intercurrent attack of hemolysis with cold associated autoimmune hemolytic anemia: A case report with
agglutinins in a case of Waldenstrom disease. Rom J Intern emphasis on pathogenic mechanisms. Am J Clin Pathol
Med 1996;34:149−154. 1996;105:567−571.
224. Vick DJ, Byrd JC, Beal CL, Chaffin DJ: Mixed-type autoim- 250. dem Borne AE, van Oers RH, Wiersinga WM, van der Tweel
mune hemolytic anemia following fludarabine treatment in a JG: Complete remission of autoimmune thrombocytopenia
120 Immune Hemolytic Anemias

after extirpation of a benign adenofibroma of the ovary. Br J 279. Arner O, Brohult J, Engstedt L, Karlson R, Sallstrom T:
Haematol 1990;74:119−120. Autoimmune hemolytic anemia in ulcerative colitis,
251. Schonitzer D, Kilga-Nogler S: Apparent “auto”-anti-DE of the cured by colectomy. Report of a case. Acta Med Scand
IgA and IgG classes in a 16-year-old girl with a mature cystic 1971;189:275−277.
ovarian teratoma. Vox Sang 1987;53:102−104. 280. Edwards FC, Truelove SC: The course and prognosis of ulcer-
252. Carreras Vescio LA, Toblli JE, Rey JA, Assaf ME, De Maria HE, ative colitis. Part III: Complications. Gut 1964;5:1−22.
Marletta J: Autoimmune hemolytic anemia associated with an 281. Gumaste V, Greenstein AJ, Meyers R, Sachar DB: Coombs-
ovarian neoplasm. Medicina (B Aires) 1983;43:415−424. positive autoimmune hemolytic anemia in ulcerative colitis.
253. Watson C: Hemolytic jaundice and macrocytic hemolytic Dig Dis Sci 1989;34:1457−1461.
anemia. Certain observations in a series of 35 cases. Ann Intern 282. Giannadaki E, Potamianos S, Roussomoustakaki M, Kyriakou
Med 1939;12:1782. D, Fragkiadakis N, Manousos ON: Autoimmune hemolytic
254. Singer K, Dameshek W: Symptomatic hemolytic anemia. Ann anemia and positive Coombs test associated with ulcerative
Intern Med 1941;15:544. colitis. Am J Gastroenterol 1997;92:1872−1874.
255. Jones E, Tilman C: A case of hemolytic anemia relieved by 283. Basista MH, Roe DC: A case presentation of hemolytic anemia
removal of an ovarian tumor. JAMA 1945;128:1225. in ulcerative colitis and review of the literature. Am J
256. Lindeboom G: Rare forms of symptomatic hemolytic anemia. Gastroenterol 1986;81:990−992.
Acta Haematol 1950;4:343. 284. Selhub J, Dhar GJ, Rosenberg IH: Inhibition of folate enzymes
257. Allibone E, Collins D: Symptomatic haemolytic anemia asso- by sulfasalazine. J Clin Invest 1978;61:221−224.
ciated with ovarian teratoma in a child. J Clin Pathol 285. van Hees PA, van Elferen LW, van Rossum JM, van Tongeren
1951;4:412. JH: Hemolysis during salicylazosulfapyridine therapy. Am J
258. De Gruchy G: The diagnosis and management of acquired Gastroenterol 1978;70:501−505.
haemolytic anaemia. Aust Ann Med 1954;3:83. 286. Altman AR, Maltz C, Janowitz HD: Autoimmune hemolytic
259. Wuhrmann F: Ginige aktuelle klinische probleme aus der anemia in ulcerative colitis: report of three cases, review of the
Humoral-pathologie. Schweiz Akad Med Wiss 1954;10:180. literature, and evaluation of modes of therapy. Dig Dis Sci
260. Andre R, Dreyfus B, Salmon C: Anemie hemolytique et kyste 1979;24:282−285.
dermoide de l’ovaire. Bull Soc Hop Paris 1955;71:2133. 287. Collazzo JA, Siddiqui MA, Deren JJ: Concomitant onset of
261. Prochazka J: Symptomaticka Hemolyticka Chudokrevnost Pri ulcerative colitis and autoimmune hemolytic anemia in an
Dermoidni Cyste Vajecniku. Vnitrni Lek 1956;2:240. 83-yr-old woman. Am J Gastroenterol 1996;91:2236−2237.
262. Muller W, Schubothe H: Symptomatische Hamolytische 288. Chochon M, Devars du Mayne JF, Meier F, Hay JM, Cerf M:
Anamie Bei Dermoidzysten. Folia Haemat 1958;2:321. Hemolytic anemia and ulcerative rectocolitis: Splenectomy or
263. Muller W, Schubothe H, Elert R: Die Hamolytische Anaemie coloproctectomy? Gastroenterol Clin Biol 1989;13:105.
als Begleiterkrankung Bei Dermoidzysten. Geburtsh 289. van Esser JW, Bos LP, Erdkamp FL: An uncommon cause of
Frauenheilk 1958;18:723. anaemia in ulcerative colitis. Neth J Med 1995;47:18−20.
264. Von Miescher P, Von Rechenberg HH, Berger J, Hollander L: 290. Fong S, Fudenberg HH, Perlman P: Ulcerative colitis with
Hamolytische anaemie bei ovarialtumor. Schweiz Med Wschr antierythrocyte antibodies. Vox Sang 1963;8:668−679.
1958;88:498. 291. Chiba M, Nakajima K, Arakawa H, Masamune O, Narisawa T:
265. Heeres P: Les anemies hemolytiques et les kystes de l’ovaire. Anti-erythrocyte antibodies in ulcerative colitis: Case report
Sem Hop Paris 1960;36:1411. and discussion on the pathophysiology of anti-erythrocyte
266. Szirmai E: Symptomatische autoantikorperbedingte Hamoly- antibody. Gastroenterol Jpn 1988;23:564−569.
tische Anamie Bei Erkrankungen der Weiblichen genitalien. 292. Yates P, Macht LM, Williams NA, Elson CJ: Red cell autoanti-
Wein Z Int Med 1961;42:320. body production by colonic mononuclear cells from a patient
267. Lorrain C, Del Solar A, Vargas Molinare R: Anemia hemolitica with ulcerative colitis and autoimmune haemolytic anaemia.
asociada a quiste dermoide del ovario. Sangre 1963;8:31. Br J Haematol 1992;82:753−756.
268. Norcross J: Hematological manifestations of malignant 293. Duhrsen U, Augener W, Zwingers T, Brittinger G: Spectrum
disease. Med Clin North Am 1963;47:345. and frequency of autoimmune derangements in lympho-
269. McAndrew G: Haemolytic anemia associated with ovarian ter- proliferative disorders: Analysis of 637 cases and comparison
atoma. Br Med J 1964;2:1307. with myeloproliferative diseases. Br J Haematol 1987;
270. Yam LT, Rudzki C, Busch S, Leithold SL: Ovarian neoplasm 67:235−239.
associated with autoimmune hemolytic anemia. Am J Obstet 294. Miller DG: The association of immune disease and malignant
Gynecol 1966;95:207−211. lymphoma. Ann Intern Med 1967;66:507−521.
271. Baker L, Brain M, Azzoopardi J, Worlledge S: Autoimmune 295. Diehl LF, Ketchum LH: Autoimmune disease and chronic
haemolytic anaemia associated with ovarian dermoid cyst. lymphocytic leukemia: Autoimmune hemolytic anemia, pure
J Clin Path 1968;21:626. red cell aplasia, and autoimmune thrombocytopenia. Semin
272. Andre R, Duhamel G, Najman A, Homberg JC, Mawas C, Oncol 1998;25:80−97.
Armangol R: Autoimmune hemolytic anemia and malignant 296. Mauro FR, Foa R, Cerretti R, et al: Autoimmune hemolytic
tumor of the ovary. Presse Med 1969;77:2133−2136. anemia in chronic lymphocytic leukemia: Clinical, therapeutic,
273. Bernstein D, Naor S, Rikover M, Menahem H: Hemolytic and prognostic features. Blood 2000;95:2786−2792.
anemia related to ovarian tumor. Obstet Gynecol 1974;43: 297. Hansen MM: Chronic lymphocytic leukaemia: Clinical studies
276−280. based on 189 cases followed for a long time. Scand J Haematol
274. Bunuel C, Val CE, Torres GM, Lapena JM: Autoimmune Suppl 1973;18:3−286.
haemolytic anaemia secondary to ovarian dermoid cyst. 298. Bartal A, Bentwich Z, Manny N, Izak G: Ethnical and clinical
Sangre (Barc) 1976;21:162−170. aspects of chronic lymphocytic leukemia in Israel: A survey on
275. Suarez C, Etcheverry M: Quiste dermoide del hilio esplenico, 288 patients. Acta Haematol 1978;60:161−171.
esplenomegalia, ictericia hemolytica y anemia grave. Arch 299. Seaman AJ, Koler RD, Pirofsky B, et al: Incidence and treat-
Argent Enf Ap Dig Nutr 1938;12:162. ment of acquired hemolytic anemia complicating chronic
276. Sandoe A: Immunohaemolytisk anaemia. Nord Med 1953;50: lymphocytic leukemia. Proceedings of the Sixth International
1565−1570. Congress of the International Society of Hematology
277. Buonanno G, Gonnella F, Pettinato G, Castaldo C: Auto- 1956:857.
immune hemolytic anemia and dermoid cyst of the mesentery. 300. Robertson TI: Complications and causes of death in B cell
A case report. Cancer 1984;54:2533−2536. chronic lymphocytic leukaemia: A long term study of
278. Lorber M, Schwartz LI, Wasserman LR: Association of anti- 105 patients. Aust N Z J Med 1990;20:44−50.
body-coated red blood cells with ulcerative colitis. Am J Med 301. De Rossi G, Granati L, Girelli G, et al: Incidence and prognos-
1955;19:887−894. tic significance of autoantibodies against erythrocytes and
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 121

platelets in chronic lymphocytic leukemia (CLL). Nouv Rev Fr 325. Guerra L, Najman A, Homberg JC, Duhamel G, Andre R:
Hematol 1988;30:403−406. Multiple autoantibodies in the course of Hodgkin’s disease:
302. Diehl LF, Bolan CD, Weiss RB: Hemolytic anemia and cancer. Interpretation of erythroblastopenia. Nouv Rev Fr Hematol
Cancer Treat Rev 1996;22:33−73. 1969;9:601−610.
303. Kyle RA, Kiely JM, Stickney JM: Acquired hemolytic anemia in 326. Decker BL, Rukes JM, et al: Hodgkin’s disease with hemolytic
chronic lymphocytic leukemia and the lymphomas. Survival anemia;complications of sternal puncture;treatment with
and response to therapy in twenty-seven cases. Arch Intern ACTH and cortisone. Amer Pract 1951;2:885−890.
Med 1959;104:929−936. 327. Blajchman MA, Gordon H: Successful pregnancy in a patient
304. Engelfriet CP, Ouwehand WH, van ‘t Veer MB, Beckers DO, with Hodgkin’s disease complicated by warm autoimmune
Maas N, von dem Borne AE. Auto-immune haemolytic hemolytic anemia and anti-e alloimmunization. Transfusion
anaemias. Bailliere’s Clin Immunol Allergy 1987;1:251−267. 1972;12:276−279.
305. Greally JF, Whelan CA, O’Connell L, O’Gorman D: Cold 328. Garratty G, Petz LD, Wallerstein RO, Fudenberg HH:
agglutinins in a case of chronic lymphatic leukaemia. A study Autoimmune hemolytic anemia in Hodgkin’s disease associ-
of the lymphocyte surface. Acta Haematol 1977;57:206−210. ated with anti-IT. Transfusion 1974;14:226−231.
306. Schwartz TB, Jager BV: Cryoglobulinemia and Raynaud’s syn- 329. Lawe JE: Successful exchange transfusion of an infant for
drome in a case of chronic lymphocytic leukemia. Cancer AIHA developing late in mother’s pregnancy. Transfusion
1949;2:319−328. 1982;22:66−68.
307. Ruzickova S, Pruss A, Odendahl M, et al: Chronic lymphocytic 330. Xiros N, Binder T, Anger B, Bohlke J, Heimpel H: Idiopathic
leukemia preceded by cold agglutinin disease: Intraclonal thrombocytopenic purpura and autoimmune hemolytic
immunoglobulin light-chain diversity in V(H)4−34 expressing anemia in Hodgkin’s disease. Eur J Haematol 1988;40:437−441.
single leukemic B cells. Blood 2002;100(9):3419−3422. 331. Brady-West DC, Thame J, West W: Autoimmune haemolytic
308. Foon KA, Rai KR, Gale RP: Chronic lymphocytic leukemia: anaemia, immune thrombocytopenia, and leucopenia. An
New insights into biology and therapy. Ann Intern Med unusual presentation of Hodgkin’s disease. West Indian Med J
1990;113:525−539. 1997;46:95−96.
309. Kipps TJ, Carson DA: Autoantibodies in chronic lymphocytic 332. Kedar A, Khan AB, Mattern JQ, Fisher J, Thomas PR, Freeman
leukemia and related systemic autoimmune diseases. Blood AI: Autoimmune disorders complicating adolescent
1993;81:2475−2487. Hodgkin’s disease. Cancer 1979;44:112−116.
310. Centola M, Lin K, Sutton C, et al: Production of anti-erythro- 333. McKenna W, Lampert I, Oakley C, Goldman J: Pel-Ebstein
cyte antibodies by leukemic and nonleukemic B cells in fever coinciding with cyclical haemolytic anaemia and
chronic lymphocytic leukemia patients. Leuk Lymphoma splenomegaly in a patient with Hodgkin’s disease. Scand J
1996;20:465−469. Haematol 1979;23:378−380.
311. Sthoeger ZM, Sthoeger D, Shtalrid M, Sigler E, Geltner D, 334. Weitberg AB, Harmon DC: Autoimmune neutropenia,
Berrebi A: Mechanism of autoimmune hemolytic hemolytic anemia, and reticulocytopenia in Hodgkin’s
anemia in chronic lymphocytic leukemia. Am J Hematol disease. Ann Intern Med 1984;100:702−703.
1993;43:259−264. 335. Chu J-Y: Autoimmmune hemolytic anemia in childnood
312. Silberstein LE, Litwin S, Carmack CE: Relationship of variable Hodgkin’s disease. American Journal of Pediatric
region genes expressed by a human B cell lymphoma secreting Hematology/Oncology 1982;4:125−128.
pathologic anti-Pr2 erythrocyte autoantibodies. J Exp Med 336. Ertem M, Uysal Z, Yavuz G, Gozdasoglu S: Immune thrombo-
1989;169:1631−1643. cytopenia and hemolytic anemia as a presenting manifestation
313. Efremov DG, Ivanovski M, Burrone OR: The pathologic of Hodgkin disease. Pediatr Hematol Oncol 2000;17:181−185.
significance of the immunoglobulins expressed by 337. Shah SJ, Warrier RP, Ode DL, Lele HE, Yu LC: Immune throm-
chronic lymphocytic leukemia B-cells in the development bocytopenia and hemolytic anemia associated with Hodgkin
of autoimmune hemolytic anemia. Leuk Lymphoma disease. J Pediatr Hematol Oncol 1996;18:227−229.
1998;28:285−293. 338. Carpentieri U, Daeschner CW, III, Haggard ME:
314. Barcellini W, Montesano R, Clerici G, et al: In vitro production Immunohemolytic anemia and Hodgkin disease. Pediatrics
of anti-RBC antibodies and cytokines in chronic lymphocytic 1982;70:320−321.
leukemia. Am J Hematol 2002;71:177−183. 339. Kalmanti M, Polychronopoulou S: Autoimmune hemolytic
315. Eisner E, Ley AB, Mayer K: Coombs’-positive hemolytic anemia anemia as an initial symptom in childhood Hodgkin’s disease.
in Hodgkin’s disease. Ann Intern Med 1967;66:258−273. Pediatr Hematol Oncol 1992;9:393−395.
316. Levine AM, Thornton P, Forman SJ, et al: Positive Coombs test 340. Garratty G, Haffleigh B, Dalziel J, Petz LD: An IgG anti-IT detec-
in Hodgkin’s disease: Significance and implications. Blood ted in a Caucasiaon American. Transfusion 1972;12:325−329.
1980;55:607−611. 341. Freedman J, Newlands M, Johnson CA: Warm IgM anti-IT
317. May RB, Bryan JH: Autoimmune hemolytic anemia and causing autoimmune haemolytic anaemia. Vox Sang 1977;
Hodgkin disease. J Pediatr 1976;89:428−429. 32:135−142.
318. Case Records of the Massachusetts General Hospital. New 342. Hafleigh EB, Wells RF, Grumet FC: Nonhemolytic IgG anti-IT.
Engl J Med 1978;298:1407−1412. Transfusion 1978;18:592−597.
319. Majumdar G: Unremitting severe autoimmune haemolytic 343. Postoway N, Capon S, Smith L, Rosenbaum D, Garratty G:
anaemia as a presenting feature of Hodgkin’s disease with Cold agglutinin syndrome caused by anti-IT: Joint Congress of
minimum tumour load. Leuk Lymphoma 1995;20:169−172. the International Society of Blood Transfusion and the
320. Bowdler AJ, Glick IW: Autoimmune hemolytic anemia as the American Association of Blood Banks Book of Abstracts. S337.
herald state of Hodgkin’s disease. Ann Intern Med 1990. (Abstract)
1966;65:761−767. 344. Ramos RR, Curtis BR, Eby CS, Ratkin GA, Chaplin H: Fatal
321. Cazenave JP, Gagnon JA, Girouard E, Bastarache A: outcome in a patient with autoimmune hemolytic anemia
Autoimmune hemolytic anemia terminating seven years later associated with an IgM bithermic anti-ITP. Transfusion
in Hodgkin’s disease. Can Med Assoc J 1973;109:748−749. 1994;34:427−431.
322. Bjorkholm M, Holm G, Merk K: Cyclic autoimmune hemolytic 345. Sallah S, Sigounas G, Vos P, Wan JY, Nguyen NP: Autoimmune
anemia as a presenting manifestation of splenic Hodgkin’s hemolytic anemia in patients with non-Hodgkin’s lymphoma:
disease. Cancer 1982;49:1702−1704. Characteristics and significance. Ann Oncol 2000;11:1571−1577.
323. Brain MC, et al: A case of malignant lymphoma with cold 346. Gronbaek K, D’Amore F, Schmidt K: Autoimmune phenomena
agglutinins. Br Med J 1969;iii:33−37. in non-Hodgkin’s lymphoma. Leuk Lymphoma 1995;18:311−316.
324. Fernandez O, Morales E, Toledo J: Autoimmune processes ter- 347. Coiffier B, Berger F, Bryon PA, Magaud JP: T-cell lymphomas:
minating 24 years later in Hodgkin’s disease. Br J Haematol Immunologic, histologic, clinical, and therapeutic analysis of
1992;81:308−309. 63 cases. J Clin Oncol 1988;6:1584−1589.
122 Immune Hemolytic Anemias

348. Horning SJ, Weiss LM, Crabtree GS, Warnke RA: Clinical and 369. Deleze M, Alarcon-Segovia D, Oria CV, et al: Hemocytopenia
phenotypic diversity of T cell lymphomas. Blood 1986; in systemic lupus erythematosus: Relationship to anti-
67:1578−1582. phospholipid antibodies. J Rheumatol 1989;16:926−930.
349. Bassan R, Pronesti M, Buzzetti M, et al: Autoimmunity and 370. Budman DR, Steinberg AD: Hematologic aspects of systemic
B-cell dysfunction in chronic proliferative disorders of lupus erythematosus: Current concepts. Ann Intern Med
large granular lymphocytes/natural killer cells. Cancer 1977;86:220−229.
1989;63:90−95. 371. Mongan ES, Leddy JP, Atwater EC, Barnett EV: Direct
350. Liaw YS, Yang PC, Su IJ, Kuo SH, Wang CH, Luh KT: Mucosa- antiglobulin (Coombs) reactions in patients with connective
associated lymphoid tissue lymphoma of the lung with cold- tissue diseases. Arthritis Rheum 1967;10:502−508.
reacting autoantibody-mediated hemolytic anemia. CHEST 372. Michael SR, Vural IL, Bassen FA, Schaefer L: The hematologic
1994;105:288−290. aspects of disseminated (systemc) lupus erythematosus. Blood
351. Economopoulos T, Stathakis N, Constantinidou M, 1951;6:1059−1072.
Papageorgiou E, Anastassiou C, Raptis S: Cold agglutinin 373. Wasserman LR, Stats D, Schwartz L, Fudenberg H:
disease in non-Hodgkin’s lymphoma. Eur J Haematol Symptomatic and hemopathic hemolytic anemia. Am J Med
1995;55:69−71. 1955;18:961−989.
352. Pascali E: Monoclonal gammopathy and cold agglutinin 374. Videbaek A: Auto-immune haemolytic anaemia in systemic
disease in non-Hodgkin’s lymphoma. Eur J Haematol lupus erythematosus. Acta Med Scand 1962;171:187−194.
1996;56:114−115. 375. Nair K, Pavithran K, Philip J, Thomas M, Geetha V: Cold
353. Costea N, Yakulis VJ, Heller P: The mechanisms of induction haemagglutinin disease in systemic lupus erythematosus.
of cold agglutinins by M: Pneumoniae. Blood 34, 829 Yonsei Med J 1997;38:233−235.
(Abstract 15), 1968. (Abstract) 376. Drenkard C, Villa AR, Alarcon-Segovia D, Perez-Vazquez ME:
354. Mainwaring CJ, Walewska R, Snowden J, et al: Fatal cold anti- Influence of the antiphospholipid syndrome in the survival of
i autoimmune haemolytic anaemia complicating hairy cell patients with systemic lupus erythematosus. J Rheumatol
leukaemia. Br J Haematol 2000;109:641−643. 1994;21:1067−1072.
355. Bar BM, Reijnders FJ, Keuning JJ, Bal H, van Beek M: Primary 377. Asherson RA, Cervera R, Piette JC, et al: Catastrophic
malignant lymphoma of the uterine cervix associated with antiphospholipid syndrome: Clues to the pathogenesis from a
cold-reacting autoantibody-mediated hemolytic anemia. Acta series of 80 patients. Medicine (Baltimore) 2001;80:355−377.
Haematol 1986;75:232−235. 378. Font J, Jimenez S, Cervera R, et al: Splenectomy for refractory
356. Isbister JP, Cooper DA, Blake HM, Biggs JC, Dixon RA, Penny Evans’ syndrome associated with antiphospholipid anti-
R: Lymphoproliferative disease with IgM lambda monoclonal bodies: Report of two cases. Ann Rheum Dis 2000;59:920−923.
protein and autoimmune hemolytic anemia: A report of four 379. Hughes GR: Thrombosis, abortion, cerebral disease, and the
cases and a review of the literature. Am J Med 1978;64: lupus anticoagulant. Br Med J (Clin Res Ed) 1983;287:
434−440. 1088−1089.
357. Sallah S, Wan JY, Hanrahan LR: Future development of lym- 380. Win N, Islam SI, Peterkin MA, Walker ID: Positive direct
phoproliferative disorders in patients with autoimmune antiglobulin test due to antiphospholipid antibodies in normal
hemolytic anemia. Clin Cancer Res 2001;7:791−794. healthy blood donors. Vox Sang 1997;72:182−184.
358. Jasty R, Strouse PJ, Castle VP: Fatal lymphoproliferative 381. Sthoeger Z, Sthoeger D, Green L, Geltner D: The role of
disease as a complication of Evans syndrome. J Pediatr anticardiolipin autoantibodies in the pathogenesis of auto-
Hematol Oncol 2000;22:460−463. immune hemolytic anemia in systemic lupus erythematosus.
359. Chaplin H, Jr. Lymphoma in primary chronic cold hemagglu- J Rheumatol 1993;20:2058−2061.
tinin disease treated with chlorambucil. Arch Intern Med 382. Hazeltine M, Rauch J, Danoff D, Esdaile JM, Tannenbaum H:
1982;142:2119−2123. Antiphospholipid antibodies in systemic lupus erythemato-
360. Otto S, Borzonyi M, Eckhardt S, Mohay A, Gergely J, Kellner sus: Evidence of an association with positive Coombs’ and
R: Immunochemical and ultrastructural investigations in a hypocomplementemia. J Rheumatol 1988;15:80−86.
patient with cold-agglutinine-active monoclonal IgMK- 383. Deleze M, Oria CV, Alarcon-Segovia D: Occurrence of both
gammopathy. Blut 1977;34:299−304. hemolytic anemia and thrombocytopenic purpura (Evans’
361. Kamiyama R, Saitoh K, Hirosawa S, Yamaguchi H, Tsukada T: syndrome) in systemic lupus erythematosus. Relationship to
Two patients with autoimmune disease developing into non- antiphospholipid antibodies. J Rheumatol 1988;15:611−615.
Hodgkin’s lymphoma. Nippon Ketsueki Gakkai Zasshi 384. Hammond A, Rudge AC, Loizou S, Bowcock SJ, Walport MJ:
1986;49:915−921. Reduced numbers of complement receptor type 1 on erythro-
362. Voulgarelis M, Kokori SI, Ioannidis JP, Tzioufas AG, Kyriaki D, cytes are associated with increased levels of anticardiolipin
Moutsopoulos HM: Anaemia in systemic lupus erythemato- antibodies. Findings in patients with systemic lupus erythe-
sus: Aetiological profile and the role of erythropoietin. Ann matosus and the antiphospholipid syndrome. Arthritis Rheum
Rheum Dis 2000;59:217−222. 1989;32:259−264.
363. Harvey AM, Shulman LE, Tumulty PA, et al: Systemic lupus 385. Cabral AR, Cabiedes J, Alarcon-Segovia D: Hemolytic anemia
erythematosus. Review of the literature and clinical analysis of related to an IgM autoantibody to phosphatidylcholine that
138 cases. Medicine 1854;33:291−437. binds in vitro to stored and to bromelain-treated human
364. Nossent JC, Swaak AJ: Prevalence and significance of haema- erythrocytes. J Autoimmun 1990;3:773−787.
tological abnormalities in patients with systemic lupus erythe- 386. Cervera R, Font J, Lopez-Soto A, et al: Isotype distribution of
matosus. Q J Med 1991;80:605−612. anticardiolipin antibodies in systemic lupus erythematosus:
365. Worlledge S: Immune haemolytic anaemias. In Hardisty RM, Prospective analysis of a series of 100 patients. Ann Rheum Dis
Weatherall DJ (eds): Blood and Its Disorders. Oxford: 1990;49:109−113.
Blackwell Scientific, 1982:479−513. 387. Alarcon-Segovia D: Pathogenetic potential of antiphospho-
366. Alger M, Alarcon-Segovia D, Rivero SJ: Hemolytic anemia and lipid antibodies. J Rheumatol 1988;15:890−893.
thrombocytopenic purpura: Two related subsets of systemic 388. Fong KY, Loizou S, Boey ML, Walport MJ: Anticardiolipin
lupus erythematosus. J Rheumatol 1977;4:351−357. antibodies, haemolytic anaemia and thrombocytopenia in
367. Ward MM, Pyun E, Studenski S: Mortality risks associated systemic lupus erythematosus. Br J Rheumatol 1992;31:
with specific clinical manifestations of systemic lupus erythe- 453−455.
matosus. Arch Intern Med 1996;156:1337−1344. 389. Arvieux J, Schweizer B, Roussel B, Colomb MG: Autoimmune
368. Alarcon-Segovia D, Deleze M, Oria CV, et al: Anti- haemolytic anaemia due to anti-phospholipid antibodies. Vox
phospholipid antibodies and the antiphospholipid syndrome Sang 1991;61:190−195.
in systemic lupus erythematosus: A prospective analysis of 500 390. Davidyuk G, Goldsby RA, Dearden MT, Stec TC,
consecutive patients. Medicine (Baltimore) 1989;68:353−365. Andrzejewski C: Autoantibodies to immature erythrocytes in
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 123

patients with the anti-phospholipid antibody syndrome. 412. Lugassy G, Reitblatt T, Ducach A, Oren S: Severe autoimmune
Transfusion 42, 103S: 2002. (Abstract) hemolytic anemia with cold agglutinin and sclerodermic
391. Del Papa N, Meroni PL, Barcellini W, et al: Antiphospholipid features⎯favorable response to danazol. Ann Hematol
antibodies cross-reacting with erythrocyte membranes: A case 1993;67:143−144.
report. Clin Exp Rheumatol 1992;10:395−399. 413. Wanchu A, Sud A, Bambery P: Linear scleroderma and
392. Lang B, Straub RH, Weber S, Rother E, Fleck M, Peter HH: autoimmune hemolytic anaemia. J Assoc Physicians India
Elevated anticardiolipin antibodies in autoimmune 2002;50:441−442.
haemolytic anaemia irrespective of underlying systemic lupus 414. Andrews J, Hall MA: Dermatomyositis-scleroderma overlap
erythematosus. Lupus 1997;6:652−655. syndrome presenting as autoimmune haemolytic anaemia.
393. Latagliata R, Celesti F, Bongarzoni V, et al: Intracardiac throm- Rheumatology (Oxford) 2002;41:956−958.
bus in a patient with autoimmune hemolytic anemia leading 415. Ellis LD, Westerman MP: Autoimmune hemolytic anemia and
to a diagnosis of antiphospholipid syndrome. Acta Haematol cancer. JAMA 1965;193:962−964.
2002;107:170−172. 416. Holland PV, Menken M, Berlin NI: Autoimmune hemolytic
394. Guzman J, Cabral AR, Cabiedes J, Pita-Ramirez L, Alarcon- anemia, acute leukemia, and carcinoma of the lung. Sodium
Segovia D: Antiphospholipid antibodies in patients with idio- phosphate P 32 treatment of polycythemia vera. Arch Intern
pathic autoimmune haemolytic anemia. Autoimmunity Med 1967;120:341−344.
1994;18:51−56. 417. Miura AB, Shibata A, Akihama T, Endo Y, Sugawara M:
395. Barker RN, Casswell KM, Reid ME, Sokol RJ, Elson CJ: Autoimmune hemolytic anemia associated with colon cancer.
Identification of autoantigens in autoimmune haemolytic Cancer 1974;33:111−114.
anaemia by a non-radioisotope immunoprecipitation method. 418. Najafi JA, Guzman LG: Tumor induced autoimmune
Br J Haematol 1992;82:126−132. hemolytic anemia in bronchogenic carcinoma: Case reports.
396. Victoria EJ, Pierce SW, Branks MJ, Masouredis SP: IgG red Mil Med 1979;144:754−756.
blood cell autoantibodies in autoimmune hemolytic anemia 419. Spira MA, Lynch EC: Autoimmune hemolytic anemia and
bind to epitopes on red blood cell membrane band 3 glycopro- carcinoma: an unusual association. Am J Med 1979;67:
tein. J Lab Clin Med 1990;115:74−88. 753−758.
397. Leddy JP, Falany JL, Kissel GE, Passador ST, Rosenfeld SI: 420. Inoue Y, Kaku K, Kaneko T, Matsumoto N: Autoimmune
Erythrocyte membrane proteins reactive with human (warm- hemolytic anemia and gastric cancer: Case report and review
reacting) anti-red cell autoantibodies. J Clin Invest of the literature. Nippon Ketsueki Gakkai Zasshi 1983;
1993;91:1672−1680. 46:836−841.
398. Galindo M, Khamashta MA, Hughes GR: Splenectomy for 421. Venzano C, De Micheli A, Cavallero GB: Autoimmune hemo-
refractory thrombocytopenia in the antiphospholipid syn- lytic anemia associated with hypernephroma. Haematologica
drome. Rheumatology (Oxford) 1999;38:848−853. 1985;70:59−61.
399. Vaughan JH, Barnett EV, Leddy JP: Autosensitivity diseases 422. Honan W, Balazs J, Jariwalla AG: Autoimmune haemolytic
Immunologic and pathogenetic concepts in lupus erythemato- anaemia (AHA) and lung carcinoma. Br J Clin Pract
sus, rheumatoid arthritis and hemolytic anemia. N Engl J Med 1986;40:35−36.
1966;275:1426−1432. 423. Girelli G, Adorno G, Perrone MP, et al: Kidney carcinoma
400. Pollock JG, Fenton E, Barrett KE: Familial autoimmune revealed by autoimmune hemolytic anemia. Haematologica
haemolytic anaemia associated with rheumatoid arthritis and 1988;73:309−311.
pernicious anaemia. Br J Haematol 1970;18:171−182. 424. Adorno G, Girelli G, Perrone MP, et al: A metastatic breast car-
401. Durance RA, Hamilton EB: Myasthenia gravis, rheumatoid cinoma presenting as autoimmune hemolytic anemia. Tumori
arthritis, vitiligo, and autoimmune haemolytic anaemia. Proc 1991;77:447−448.
R Soc Med 1971;64:61−62. 425. Hibino S, Stoller RG, Jacobs SA: Autoimmune haemolytic
402. Chapman AH: Concurrent development of acute autoimmune anaemia associated with transitional cell carcinoma. Lancet
haemolytic anaemia with rheumatoid arthritis. Proc R Soc 1992;340:373.
Med 1972;65:1013−1015. 426. Nakao A, Iwagaki H, Notohara K, et al: Successful resection of
403. Suda T, Oike S, Omine M, Naruse T, Tsuchiya J: Haemolytic rectal carcinoma in an Evans’ syndrome patient followed by
anaemia due to warm type autohaemagglutin in a patient with predonisolone and high-dose immunoglobulin: Report of a
rheumatoid arthritis. Nippon Ketsueki Gakkai Zasshi case. Acta Med Okayama 2001;55:253−257.
1977;40:466−471. 427. Lands R, Foust J: Renal cell carcinoma and autoimmune
404. Maharaj D: Autoimmune haemolytic anaemia associated with hemolytic anemia. South Med J 1996;89:444−445.
rheumatoid arthritis and paroxysmal nocturnal haemoglobin- 428. Kamra D, Boselli J, Sloane BB, Gladstone DE: Renal cell
uria. Acta Haematol 1986;75:241. carcinoma induced Coombs negative autoimmune hemolytic
405. Chaves FC, Rodrigo FG, Franco ML, Esteves J: Systemic scle- anemia and severe thrombocytopenia responsive to nephrec-
rosis associated with auto-immune haemolytic anaemia. Br J tomy. J Urol 2002;167:1395.
Dermatol 1970;82:298−302. 429. Cao L, Kaiser P, Gustin D, Hoffman R, Feldman L: Cold agglu-
406. Rosenthal DS, Sack B: Autoimmune hemolytic anemia in scle- tinin disease in a patient with uterine sarcoma. Am J Med Sci
roderma. JAMA 1971;216:2011−2012. 2000;320:352−354.
407. Ivey KJ, Hwang YF, Sheets RF: Scleroderma associated with 430. Gordon PA, Baylis PH, Bird GW: Tumour-induced
thrombocytopenia and Coombs-positive hemolytic anemia. autoimmune haemolytic anaemia. Br Med J 1976;1:1569−1570.
Am J Med 1971;51:815−817. 431. Bradley GW, Harvey M: Haemolytic anaemia with hyper-
408. Loft B, Olsen F: Autoimmune haemolytic anaemia with posi- nephroma. Postgrad Med J 1981;57:46−47.
tive Ham and Crosby’s test and scleroderma. A case report. 432. Kornberg A, Naparstek E, Hershko C: Cryopathic haemolytic
Scand J Haematol 1973;11:131−134. anaemia associated wtih uterus myomatosus. Acta Haematol
409. Sumithran E: Progressive systemic sclerosis and 1980;63:235.
autoimmune haemolytic anaemia. Postgrad Med J 433. Johnson P, Gualtieri RJ, Mohler DN, Carpenter JT: Auto-
1976;52:173−176. immune hemolytic anemia associated with a hypernephroma.
410. Jones E, Jones JV, Woodbury JF, Carr RI, Skanes V: Scleroderma South Med J 1985;78:1129−1131.
and hemolytic anemia in a patient with deficiency of IgA and 434. Wortman J, Rosse W, Logue G: Cold agglutinin autoimmune
C4: A hitherto undescribed association. J Rheumatol hemolytic anemia in nonhematologic malignancies. Am J
1987;14:609−612. Hematol 1979;6:275−283.
411. Pettersson T, von Bonsdorff M: Auto-immune haemolytic 435. Chen RT, Pless R, Destefano F: Epidemiology of autoim-
anaemia and thrombocytopenia in scleroderma. Acta mune reactions induced by vaccination. J Autoimmun
Haematol 1988;80:179−180. 2001;16:309−318.
124 Immune Hemolytic Anemias

436. Cohen AD, Shoenfeld Y: Vaccine-induced autoimmunity. 459. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Cold-
J Autoimmun 1996;9:699−703. antibody syndromes III: Haemolytic anaemia following
436a. Offit PA, Hackett CJ: Addressing parents’ concerns: Do mycoplasma pneumonia. In The Haemolytic Anaemias, 3rd
vaccines cause allergic or autoimmune diseases? Pediatrics ed. vol. 3, The Auto-Immune Haemolytic Anaemias. New
2003;111:653–659. York: Churchill Livingstone, 1992: 296−312.
437. Kolbl H: Klinik und Therapie der akuten erworbenen 460. Linz DH, Tolle SW, Elliot DL: Mycoplasma pneumoniae pneu-
hamolytischen Anamien im Kindesalter. Ostdeutsch Z monia. Experience at a referral center. West J Med 1984;
Kinderheilk 1955;11:27. 140:895−900.
438. Dameshek W, Schwartz R: Hemolytic mechanisms. Ann NY 461. Smith GN, Weir WR: Cold agglutinins accompanying
Acad Sci 1959;77:589. Mycoplasma pneumoniae infection. Br Med J 1980;281:
439. Gedikoglu AG, Cantez T: Haemolytic-anaemia relapses after 1391−1392.
immunisation and pertussis. Lancet 1967;2:894−895. 462. Platt WR, Ward DS: Cold isohemagglutinins. Their association
440. Bossi E, Wagner HP: Autoimmune hemolytic anemia and with hemolytic anemia and multiple thromboses in primary
cytomegalovirus infection in a six- months-old child, treated atypical pneumonia: A brief review of the clinical and labora-
with azathioprine. Helv Paediatr Acta 1972;27:155−162. tory problems involved. Am J Clin Path 1945;15:202−209.
441. Haneberg B, Matre R, Winsnes R, Dalen A, Vogt H, Finne PH: 463. Carey RM, Wilson JL, Tamerin JA: Gangrene of feet and
Acute hemolytic anemia related to diphtheria-pertussis- hemolytic anemia associated with cold hemagglutinins in
tetanus vaccination. Acta Paediatr Scand 1978;67:345−350. atypical pneumonia. Harlem Hosp Bull 1948;1:25−32.
442. Johnson ST, McFarland JG, Casper JT, et al: An unusual case of 464. Stats D, Wasserman LR, Rosenthal N: Hemolytic anemia with
autoimmune hemolytic anemia following DPT vaccination. hemoglobinuria. Amer J Clin Path 1948;18:757−777.
Transfusion 1989;29:50S. 465. Kumar S, Singh MM, Bhatia BB: Symmetrical peripheral gan-
443. Downes KA, Domen RE, McCarron KF, Bringelsen KA: Acute grene in acquired hemolytic anemia. Acta haemat (Basel)
autoimmune hemolytic anemia following DTP vaccination: 1958;19:369−377.
Report of a fatal case and review of the literature. Clinical 466. Schulman P, Piemonte TC, Singh B: Acute renal failure,
Pediatrics 2001;40:355−358. hemolytic anemia, and Mycoplasma pneumoniae. JAMA
444. Seltsam A, Shukry-Schulz S, Salama A: Vaccination-associated 1980;244:1823−1824.
immune hemolytic anemia in two children. Transfusion 467. Lawson DH, Lindsay RM, Sawers JD, et al: Acute renal failure
2000;40:907−909. in the cold-agglutination syndrome. Lancet 1968;2:704−705.
445. Martinez E, Domingo P: Evans’s syndrome triggered by 468. Shibasaki T, Gomi H, Ohno I, Ishimoto F, Sakai O: A case of
recombinant hepatitis B vaccine. Clin Infect Dis 1992;15:1051. chronic renal failure followed by cold agglutinin due to
446. Howson CP, Fineberg HV: Adverse events following pertussis Mycoplasma pneumoniae infection. Nephron 1991;57:249−250.
and rubella vaccines. Summary of a report of the Institute of 469. Poth JL, Sharp GS, Schrier SL: Cold agglutinin disease and the
Medicine. JAMA 1992;267:392−396. nephrotic syndrome. JAMA 1970;211:1989−1992.
447. Berkowitz FE: Hemolysis and infection: Categories and 470. Cherry JD: Anemia and mucocutaneous lesions due to
mechanisms of their interrelationship. Rev Infect Dis Mycoplasma pneumoniae infections. Clin Infect Dis 1993;17
1991;13:1151−1162. (Suppl. 1):S47–S51.
448. Seitz RC, Buschermohle G, Dubberke G, Herbrand R, 471. Chu CS, Braun SR, Yarbro JW, Hayden MR: Corticosteroid
Maiwald M, Hellwege HH: The acute infection-associated treatment of hemolytic anemia associated with Mycoplasma
hemolytic anemia of childhood: Immunofluorescent detection pneumoniae pneumonia. South Med J 1990;83:1106−1108.
of microbial antigens altering the erythrocyte membrane. Ann 472. Tsuruta R, Kawamura Y, Inoue T, Kasaoka S, Sadamitsu D,
Hematol 1993;67:191−196. Maekawa T: Corticosteroid therapy for hemolytic anemia and
449. Morrow WJ, Isenberg DA, Sobol RE, Stricker RB, Kieber- respiratory failure due to Mycoplasma pneumoniae pneumo-
Emmons T: AIDS virus infection and autoimmunity: A per- nia. Intern Med 2002;41:229−232.
spective of the clinical, immunological, and molecular origins 473. Chan LY, Chan JC: Mycoplasma pneumoniae infection pre-
of the autoallergic pathologies associated with HIV disease. senting as haemolytic anaemia. Br J Hosp Med 1997;58:170.
Clin Immunol Immunopathol 1991;58:163−180. 474. Fink FM, Dengg K, Kilga-Nogler S, Schonitzer D, Berger H:
450. Meite M, Leonard S, Idrissi ME, Izui S, Masson PL, Coutelier Cold haemagglutinin disease complicating Mycoplasma
JP: Exacerbation of autoantibody-mediated hemolytic anemia pneumoniae infection in a child under cytotoxic cancer treat-
by viral infection. J Virol 2000;74:6045−6049. ment. Eur J Pediatr 1992;151:435−437.
451. Konig AL, Schabel A, Sugg U, Brand U, Roelcke D: 475. Tanowitz HB, Robbins N, Leidich N: Hemolytic anemia.
Autoimmune hemolytic anemia caused by IgG lambda- Associated with severe Mycoplasma pneumoniae pneumo-
monotypic cold agglutinins of anti-Pr specificity after rubella nia. N Y State J Med 1978;78:2231−2232.
infection. Transfusion 2001;41:488−492. 476. Maisel JC, Babbitt LH, John TJ: Fatal Mycoplasma pneumo-
452. Murray HW, Masur H, Senterfit LB, Roberts RB: The protean niae infection with isolation of organisms from lung. JAMA
manifestations of Mycoplasma pneumoniae infection in 1967;202:287−290.
adults. Am J Med 1975;58:229−242. 477. Louie EK, Ault KA, Smith BR, Hardman EL, Quesenberry PJ:
453. Chanock RM: Mycoplasma infections of man. N Engl J Med IgG-mediated haemolysis masquerading as cold agglutinin-
1965;273:1257−1264. induced anaemia complicating severe infection with
454. Denny FW, Clyde WA, Jr, Glezen WP: Mycoplasma pneumo- mycoplasma pneumoniae. Scand J Haematol 1985;35:264−269.
niae disease: Clinical spectrum, pathophysiology, epidemiol- 478. Schmidt PJ, Barile MF, McGinniss MH: Mycoplasma
ogy, and control. J Infect Dis 1971;123:74−92. (Pleuropneumonia-like organismns) and blood group I:
455. Daxbock F, Zedtwitz-Liebenstein K, Burgmann H, Graninger W: Associations with neoplastic disease. Nature (Lond) 1965;205:
Severe hemolytic anemia and excessive leukocytosis masking 371−372.
mycoplasma pneumonia. Ann Hematol 2001;80:180−182. 479. Smith CB, McGinniss MH, Schmidt PJ: Changes in erythrocyte
456. Ali NJ, Sillis M, Andrews BE, Jenkins PF, Harrison BD: The I agglutinogen and anti-I-agglutinins during Mycoplasma
clinical spectrum and diagnosis of Mycoplasma pneumoniae pneumoniae infection in man. J Immunol 1967;99:333−339.
infection. Q J Med 1986;58:241−251. 480. Feizi T, Taylor-Robinson D, Shields MD, Carter RA:
457. Feizi T: Cold agglutinins, the direct coombs’ test and serum Production of cold agglutinins in rabbits immunized with
immunoglobulins in Mycoplasma pneumoniae infection. Ann human erythrocytes treated with Mycoplasma pneumoniae.
N Y Acad Sci 1967;143:801−812. Nature 1969;222:1253−1256.
458. Jacobson LB, Longstreth GF, Edgington TS: Clinical and 481. Loomes LM, Uemura K, Childs RA, et al: Erythrocyte recep-
immunologic features of transient cold agglutinin-hemolytic tors for Mycoplasma pneumoniae are sialylated oligosaccha-
anemia. Am J Med 1973;54:514−521. rides of Ii antigen type. Nature 1984;307:560−563.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 125

482. Costea N, Yakulis V, Heller P: Experimental production of 504. Godal HC, Skaga E: Aggravation of congenital spherocytosis
cold agglutinins in rabbits. Blood 1965;26:323−339. during infectious mononucleosis. Scand J Haematol 1969;6:
483. Costea N, Yakulis VJ, Heller P: Inhibition of cold agglutinins 33−35.
(anti-I) by M: pneumoniae antigens. Proc Soc Exp Biol Med 505. Gehlbach SH, Cooper BA: Haemolytic anaemia in infectious
1972;139:476−479. mononucleosis due to inapparent congenital spherocytosis.
484. Lind K: Production of cold agglutinins in rabbits induced by Scand J Haematol 1970;7:141−144.
Mycoplasma pneumoniae, Listeria monocytogenes or 506. Taylor JJ: Haemolysis in infectious mononucleosis: Inapparent
Streptococcus MG. Acta Pathol Microbiol Scand [B] Microbiol congenital spherocytosis. Br Med J 1973;4:525−526.
Immunol 1973;81:487−496. 507. Jenkins WJ, Koster HG, Marsh WL, Carters RL: Infectious
485. Janney FA, Lee LT, Howe C: Cold hemagglutinin cross-reac- mononucleosis: An unsuspected source of anti-i. Brit J
tivity with Mycoplasma pneumoniae. Infect Immun 1978;22: Haemat 1965;11:480−483.
29−33. 508. Calvo RS, Stein W, Kochwa S, Rosenfield RE: Acute hemolytic
486. Bartholomew JR, Bell WR, Shirey RS: Cold agglutinin anemia due to anti-i: Frequent cold agglutinins in infectious
hemolytic anemia: Management with an environmental suit. mononucleosis. Journal of Clinical Investigation 44, 1033.
Ann Intern Med 1987;106:243−244. 1965. (Abstract)
486a. Ness PM, Bell WR, Shirey RS: Transfusion medicine illus- 509. Rosenfield RE, Schmidt PJ, Calvo RC, McGinniss MH: Anti-i,
trated. Novel management of cold agglutinin disease. Trans- a frequent cold agglutinin in infectious mononucleosis. Vox
fusion 2003;43:839. Sang 1965;10:631−634.
487. Turtzo DF, Ghatak PK: Acute hemolytic anemia with Myco- 510. Wollheim FA, Williams RC, Jr: Studies on the macroglobulins
plasma pneumoniae pneumonia. JAMA 1976;236:1140−1141. of human serum. I: Polyclonal immunoglobulin class M (IgM)
488. Lindstrom FD, Stahl-Furenhed B: Autoimmune haemolytic increase in infectious mononucleosis. N Engl J Med
anaemia complicating Mycoplasma pneumoniae infection. 1966;274:61−67.
Scand J Infect Dis 1981;13:233−235. 511. Troxel DB, Innella F, Cohen RJ: Infectious mononucleosis com-
489. Helms CM: Infections caused by Mycoplasma pneumoniae plicated by hemolytic anemia due to anti-i. Am J Clin Pathol
and the genital mycoplasmas. In: Humes HD, et al (eds): 1966;46:625−631.
Kelly’s Textbook of Internal Medicine. New York: Lippincott 512. Burkart PT, Hsu TC: IgM cold-warm hemolysins in infectious
Williams & Wilkins, 2000:2110−2113. mononucleosis. Transfusion 1979;19:535−538.
490. Einzig MJ, Neerhout RC: Hemolytic anemia in infectious 513. Gronemeyer P, Chaplin H, Ghazarian V, Tuscany F, Wilner
mononucleosis. Clin Pediatr (Phila) 1969;8:171−173. GD: Hemolytic anemia complicating infectious mononucleo-
491. Hoagland RJ: Infectious Mononucleosis. New York: Grune & sis due to the interaction of an IgG cold anti-i and an IgM cold
Stratton, 1967. rheumatoid factor. Transfusion 1981;21:715−718.
492. Boughton CR: Glandular fever. A study of a hospital series in 514. Wilkinson LS, Petz LD, Garratty G: Reappraisal of the role of
Sydney. Med J Aust 1970;2:529−535. anti-i in haemolytic anaemia in infectious mononucleosis. Br J
493. Pagano JS: Epstein-Barr virus and the infectious mononucleo- Haematol 1973;25:715−722.
sis syndrome. In: Humes HD (ed): Kelly’s Textbook of Internal 515. Lee CH, Hagen MA, Chong BH, Grace CS, Rozenberg MC:
Medicine. Philadelphia: Lippincott Williams & Wilkins, The Lewis system and secretor status in autoimmune
2000:2179−2185. hemolytic anemia complicating infectious mononucleosis.
494. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Cold- Transfusion 1980;20:585−588.
antibody syndromes IV: Haemolytic anaemia following 516. Ellis LB, Wollenman OJ, Stetson RP: Autohemagglutinins and
infectious mononucleosis and other viral infections. In The hemolysins with hemoglobinuria and acute hemolytic
Haemolytic Anaemias, 3rd ed. vol. 3, The Auto-Immune anaemia, in an illness resembling infectious mononucleosis.
Haemolytic Anaemias. New York: Churchill Livingstone, Blood 3, 419−429. 1948. (Abstract)
1992:313−328. 517. Perkin RL, Fox AD, Richards WL, King MH: Acute hemolytic
495. Woodruff RK, McPherson AJ: Severe haemolytic anaemia anemia secondary to infectious mononucleosis. Can Med
complicating infectious mononucleosis. Aust N Z J Med Assoc J 1979;121:1095−1097.
1976;6:569−570. 518. Zuelzer WW, Stulberg CS, Page RH, Teruya J, Brough AJ:
496. Worlledge SM, Dacie JV: Haemolytic and other anaemias in Etiology and pathogenesis of acquired hemolytic anemia.
infectious mononucleosis. In: Carter RL, Penman HG (eds): Transfusion 1966;6:438−461.
Infectious Mononucleosis: Oxford: Blackwell Scientific, 519. Zuelzer WW, Mastrangelo R, Stulberg CS, Poulik MD, Page
1969:82−98. RH, Thompson RI: Autoimmune hemolytic anemia: Natural
497. Silber M, Richards JD, Jacobs P: Life-threatening haemolytic history and viral-immunologic interactions in childhood. Am
anaemia and infectious mononucleosis: A case report. S Afr J Med 1970;49:80−93.
Med J 1985;67:183−185. 520. Kantor GL, Goldberg LS, Johnson BL, Jr, Derechin MM,
498. Tonkin AM, Mond HG, Alford FP, Hurley TH: Severe acute Barnett EV: Immunologic abnormalities induced by post-
haemolytic anaemia complicating infectious mononucleosis. perfusion cytomegalovirus infection. Ann Intern Med
Med J Aust 1973;2:1048−1050. 1970;73:553−558.
499. Bowman HS, Marsh WL, Schumacher HR, Oyen R, Reihart J: 521. Harris AI, Meyer RJ, Brody EA: Cytomegalovirus-induced
Auto anti-N immunohemolytic anemia in infectious mononu- thrombocytopenia and hemolysis in an adult. Ann Intern Med
cleosis. Am J Clin Pathol 1974;61:465−472. 1975;83:670−671.
500. Palanduz A, Yildirmak Y, Telhan L, et al: Fulminant hepatic 522. van Spronsen DJ, Breed WP: Cytomegalovirus-induced
failure and autoimmune hemolytic anemia associated with thrombocytopenia and haemolysis in an immunocompetent
Epstein-Barr virus infection. J Infect 2002;45:96−98. adult. Br J Haematol 1996;92:218−220.
501. Young LE, Izzo MJ, Platzer RF: Hereditary spherocytosis. 523. Salloum E, Lundberg WB: Hemolytic anemia with positive
I: Clinical hematologic and genetic features in 28 cases, with direct antiglobulin test secondary to spontaneous cytomegalo-
particular reference to the osmotic and mechanical fragility of virus infection in healthy adults. Acta Haematol 1994;92:39−41.
incubated erythrocytes. Blood 1951;6:1073−1098. 524. Coombs RR: Cytomegalic inclusion-body disease associated
502. Bean RHD: Haemolytic anaemia complicating infectious with acquired autoimmune haemolytic anaemia. Br Med J
mononucleosis, with report of a case. Med J Aust 1957;1: 1968;2:743−744.
386−389. 525. Houston MC, Porter LL, III, Jenkins DE, Jr, Flexner JM: Acute
503. DeNardo GL, Ray JP: Hereditary spherocytosis and infectious immune hemolytic anemia in adults after cytomegalovirus
mononucleosis, with acquired hemolytic anemia: Report of a infection. South Med J 1980;73:1270−1274.
case and review of the literature. Amer J Clin Path 1963; 526. Horwitz CA, Henle W, Henle G, et al: Clinical and laboratory
39:284−288. evaluation of cytomegalovirus-induced mononucleosis in pre-
126 Immune Hemolytic Anemias

viously healthy individuals: Report of 82 cases. Medicine immunodeficiency-virus (HIV) infection. Br J Haematol
(Baltimore) 1986;65:124−134. 1987;67:109−114.
527. Horwitz CA, Skradski K, Reece E, et al: Haemolytic anaemia in 551. Scadden DT, Zon LI, Groopman JE: Pathophysiology and man-
previously healthy adult patients with CMV infections: Report agement of HIV-associated hematologic disorders. Blood
of two cases and an evaluation of subclinical haemolysis in 1989;74:1455−1463.
CMV mononucleosis. Scand J Haematol 1984;33:35−42. 552. Saif MW: HIV-associated autoimmune hemolytic anemia: An
528. Gavazzi G, Leclercq P, Bouchard O, Bosseray A, Morand P, update. AIDS Patient Care STDS 2001;15:217−224.
Micoud M: Association between primary cytomegalovirus 553. Schreiber ZA, Loh SH, Charles N, Abeebe LS: Autoimmune
infection and severe hemolytic anemia in an immunocompe- hemolytic anemia in patients with the acquired immuno-
tent adult. Eur J Clin Microbiol Infect Dis 1999;18:299−301. deficiency syndrome (AIDS). Blood 62, 117A: 1983. (Abstract)
529. Eddleston M, Peacock S, Juniper M, Warrell DA: Severe 554. Miller KD, Gralnik HR, Rick ME: Hematologic problems in
cytomegalovirus infection in immunocompetent patients. Clin patients with AIDS and ARC: The NIH experience. Blood 70,
Infect Dis 1997;24:52−56. 124A: 1987. (Abstract)
530. Dietz AJ, Jr: Cytomegalovirus infection with carditis, hepatitis, 555. Puppo F, Torresin A, Lotti G, Balleari E, Orlando G, Indiveri F:
and anemia. Postgrad Med 1981;70:203−208. Autoimmune hemolytic anemia and human immunodeficiency
531. Berlin BS, Chandler R, Green D: Anti-”i” antibody and hemo- virus (HIV) infection. Ann Int Med 1988;109:249−250.
lytic anemia associated with spontaneous cytomegalovirus 556. Rapoport AP, Rowe JM, McMican A: Life-threatening autoim-
mononucleosis. Am J Clin Pathol 1977;67:459−461. mune hemolytic anemia in a patient with the acquired
532. Reller LB: Granulomatous hepatitis associated with acute immune deficiency syndrome. Transfusion 1988;28:190−191.
cytomegalovirus infection. Lancet 1973;7793.:20−22. 557. Gaffuri L, Repetto L, Rossi E, Oliva C, Rosso R, Rizzo F:
533. Aguado JM, Castrillo JM, Sanz J, Serrano J: Severe haemolytic Haemolytic anaemia with positive cryoglobulin test in a HIV
anaemia due to cold anti-”i” antibodies associated with positive man. Eur J Cancer 1991;27:304.
cytomegalovirus infection. Postgrad Med J 1990;66:392−394. 558. Taneja-Uppal N, Rappaport S, Berger BJ, Davidson E, Rahal JJ:
534. McCarthy LJ, Danielson CF, Fernandez C, et al: Intensive Human immunodeficiency virus (HIV) infection and
plasma exchange for severe autoimmune hemolytic anemia in hemolytic anemia. Ann Intern Med 1989;111:340−341.
a four-month-old infant. J Clin Apheresis 1999;14:190−192. 559. Tongol JM, Gounder MP, Butala A, Rabinowitz M: HIV-related
535. Murray JC, Bernini JC, Bijou HL, Rossmann SN, Mahoney DH, autoimmune hemolytic anemia: Good response to zidovudine.
Jr, Morad AB: Infantile cytomegalovirus-associated autoimmune J Acquir Immune Defic Syndr 1991;4:1163−1164.
hemolytic anemia. J Pediatr Hematol Oncol 2001;23:318−320. 560. Pan A, Pirola F, Bergonzi C, Petrini C, Carnevale G:
536. Hausler M, Schaade L, Hutschenreuter G, Hannig U, Reticulocytopenic immune hemolytic anemia in an AIDS
Kusenbach G: Severe cytomegalovirus-triggered autoimmune patient. Int Conf AIDS 1992;8:120.
hemolytic anemia complicating vertically acquired HIV infec- 561. Romo M, Martin VC, Guerra JL, Marinas C, de Diego T: IgM
tion. Eur J Clin Microbiol Infect Dis 2000;19:57−60. cryoagglutinin with biphasic hemolysin activity in a patient
537. Telen MJ, Roberts KB, Bartlett JA: HIV-associated autoimmune with AIDS: Sangre (Barc) 1991;36:165−166.
hemolytic anemia: Report of a case and review of the litera- 562. Lopez Dupla JM, Rodriguez PA, Martinez MP, de Castro CJ,
ture. J Acquir Immune Defic Syndr 1990;3:933−937. Lavilla UP, Gil AA: Hemolytic anemia due to cold-reacting
538. Kreuzer KA, Rockstroh JK: Pathogenesis and pathophysiology antibodies: association with human immunodeficiency virus
of anemia in HIV infection. Ann Hematol 1997;75:179−187. infection and non-Hodgkin’s lymphoma. Med Clin (Barc)
539. Claster S: Biology of anemia, differential diagnosis, and treat- 1992;98:502−504.
ment options in human immunodeficiency virus infection. 563. Dollus C, Dalle JH, Reviron M, Leverger G, Lasfargues G,
J Infect Dis 2002;185 (Suppl. 2):S105–S109. Courpotin C: Pediatric case of HIV associated autoimmune
540. Coyle TE: Hematologic complications of human immun- hemolytic anemia. Int Conf AIDS 1993;9:450.
odeficiency virus infection and the acquired immuno- 564. Sukthankar AD, Bowman CA, Carey M, Radcliffe KW: HIV
deficiency syndrome. Med Clin North Am 1997;81:449−470. infection with haemolytic anaemia. Genitourin Med
541. Hambleton J: Hematologic complications of HIV infection. 1997;73:66−69.
Oncology (Huntingt) 1996;10:671−680. 565. Borgna-Pignatti C, Bezzi TM, Reverberi R: Fatal ceftriaxone-
542. Toy PT, Reid ME, Burns M: Positive direct antiglobulin test induced hemolysis in a child with acquired immunodeficiency
associated with hyperglobulinemia in acquired immuno- syndrome. Pediatr Infect Dis J 1995;14:1116−1117.
deficiency syndrome (AIDS). Am J Hematol 1985;19:145−150. 566. Prazuck T, Semaille C, Roques S: Fatal acute haemolysis in an
543. McGinniss MH, Macher AM, Rook AH, Alter HJ: Red cell AIDS patient treated with indinavir. AIDS 1998;12:531−533.
autoantibodies in patients with acquired immune deficiency 567. Capron L, Kim YU, Laurian C, Bruneval P, Fiessinger JN:
syndrome. Transfusion 1986;26:405−409. Atheroembolism in HIV-positive individuals. Lancet
544. Zon LI, Arkin C, Groopman JE: Haematologic manifestations 1992;340:1039−1040.
of the human immune deficiency virus (HIV). Br J Haematol 568. Pinilla J, Hill AR: Thromboembolism associated with acquired
1987;66:251−256. immunodeficiency syndrome. CHEST 1992;102:1634.
545. Bordin JO, Kerbauy J, Souza-Pinto JC, et al: Quantitation of red 569. Pulik M, Lionnet F, Couderc LJ, Matheron S, Saimot AG:
cell-bound IgG by an enzyme-linked antiglobulin test in Thromboembolic disease and human immunodeficiency virus
human immunodeficiency virus-infected persons. Transfusion infection. Blood 1993;82:2931.
1992;32:426−429. 570. Cohen JR, Lackner R, Wenig P, Pillari G: Deep venous
546. De Angelis V, Biasinutto C, Pradella P, Vaccher E, Spina M, thrombosis in patients with AIDS: N Y State J Med
Tirelli U: Clinical significance of positive direct antiglobulin 1990;90:159−161.
test in patients with HIV infection. Infection 1994;22:92−95. 571. Saif MW, Bona R, Greenberg B: AIDS and thrombosis:
547. Inada Y, Lange M, McKinley GF, et al: Hematologic correlates Retrospective study of 131 HIV-infected patients. AIDS Patient
and the role of erythrocyte CR1 (C3b receptor) in the develop- Care STDS 2001;15:311−320.
ment of AIDS. AIDS Res 1986;2:235−247. 572. Haemolytic transfusion reactions. In: Mollison PL, Engelfriet
548. Simpson MB, Delong N: Autoimmune hemolytic anemia in a CP, Contreras M (eds): Blood Transfusion in Clinical Medicine,
patient with acquired immune deficiency syndrome. Blood 70, 10th ed. Oxford: Blackwell Science Ltd, 1997:358−389.
127A: 1987. (Abstract) 573. Ries M, Simon S, Hummer HP, Leipold G: Acute mesenteric
549. Lepennec PY, Lefrere JJ, Rouzaud AM, Rouger P: Red cell vein thrombosis. A rare complication after splenectomy due to
autoantibodies in asymptomatic HIV-infected subjects. autoimmune hemolytic anemia in childhood. Monatssch
Transfusion 1989;29:465−466. Kinderheilkd 1993;141:779−781.
550. van der Lelie J, Lange JM, Vos JJ, van Dalen CM, Danner SA, 574. Terada K, Tanaka H, Mori R, Kataoka N, Uchikawa M:
dem Borne AE: Autoimmunity against blood cells in human Hemolytic anemia associated with cold agglutinin during
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 127

chickenpox and a review of the literature. J Pediatr Hematol virus infection: Clinical description of 35 cases. Medicine
Oncol 1998;20:149−151. (Baltimore) 2003;82:87–96.
575. Borbolla L, Chediak B: Anemia hemolitica de etiologia viral 598. Chao TC, Chen CY, Yang YH, Chen PM, Chang FY, Lee SD:
revision de la literature. Rev Cubana Pediatr 1953;25: Chronic hepatitis C virus infection associated with primary
557−564. warm-type autoimmune hemolytic anemia. J Clin Gastro-
576. Friedman HD, Dracker RA: Cold agglutinin disease after enterol 2001;33:232−233.
chicken pox. An uncommon complication of a common 599. Srinivasan R: Autoimmune hemolytic anemia in treatment-
disease. Am J Clin Pathol 1992;97:92−96. naive chronic hepatitis C infection. J Clin Gastroenterol
577. Herron B, Roelcke D, Orson G, Myint H, Boulton FE: Cold 2001;32:245−247.
autoagglutinins with anti-Pr specificity associated with fresh 600. Fellermann K, Stange EF: Chronic hepatitis C, common vari-
varicella infection. Vox Sang 1993;65:239−242. able immunodeficiency and autoimmune hemolytic anemia.
578. Johnson AM: Cold agglutinin disease after chickenpox. Am J Coincidence by chance or common etiology? Hepato-
Clin Pathol 1992;98:271−272. gastroenterology 2000;47:1422−1424.
579. Northoff H, Martin A, Roelcke D: An IgG kappa-monotypic 601. Landau A, Castera L, Buffet C, Tertian G, Tchernia G: Acute
anti-Pr 1h associated with fresh varicella infection. Eur J autoimmune hemolytic anemia during interferon-alpha
Haematol 1987;38:85−88. therapy for chronic hepatitis C. Dig Dis Sci 1999;44:1366−1367.
580. Parashar S, Bhandary S, Menezes S, et al: Auto immune 602. Kazuta Y, Watanabe N, Sagawa K, et al: A case of autoimmune
haemolytic anaemia. J Assoc Physicians India 1981;29:679−682. hemolytic anemia induced by IFN-beta therapy for type-C
581. Kaiser AD, Bradford WL: Severe hemoglobinuria in a child chronic hepatitis. Fukushima J Med Sci 1995;41:43−49.
occurring in the prodromal stage of chickenpox. Arch Pediatr 603. Hizawa N, Kojima J, Kojima T, et al: A patient with chronic hep-
1929;46:571−577. atitis C who simultaneously developed interstitial pneumonia,
582. Sanchis CJ, Carbonell UF: Autoimmune hemolytic anemia hemolytic anemia and cholestatic liver dysfunction after alpha-
with anti-DC specificity following a primary infection by interferon administration. Intern Med 1994;33:337−341.
Varicella virus. Haematologica 1997;824:508−509. 604. Hirashima N, Mizokami M, Orito E, et al: Chronic hepatitis C
583. Ziebold C, von Kries R, Lang R, Weigl J, Schmitt HJ: Severe complicated by Coombs-negative hemolytic anemia during
complications of varicella in previously healthy children in interferon treatment. Intern Med 1994;33:300−302.
Germany: A 1-year survey. Pediatrics 2001;108:E79. 605. Moccia F, Tognoni E, Boccaccio P: Autoimmune hemolytic
584. Kadota Y, Fujinami S, Tagawa Y, et al: Haemolytic anaemia anemia in chronic hepatitis C virus infection: An unusual
caused by anti-Pra following rubella infection. Transfusion extrahepatic autoimmune manifestation. Ann Ital Med Int
Med 1993;3:207−209. 2001;16:256−259.
585. Ueda K, Shingaki Y, Sato T, Tokugawa K, Sasaki H: Hemolytic 606. Longo F, Hastier P, Buckley MJ, Chichmanian RM, Delmont JP:
anemia following postnatally acquired rubella during the Acute hepatitis, autoimmune hemolytic anemia, and erythrob-
1975–1977 rubella epidemic in Japan. Clin Pediatr (Phila) lastocytopenia induced by ceftriaxone. Am J Gastroenterol
1985;24:155−157. 1998;93:836−837.
586. Miyazaki S, Ohtsuka M, Ueda K, Shibata R, Goya N: Coombs 607. Kondo H, Kajii E, Oyamada T, Kasahara Y: Direct antiglobulin
positive hemolytic anemia in congenital rubella. J Pediatr test negative autoimmune hemolytic anemia associated with
1979;94:759−760. autoimmune hepatitis. Int J Hematol 1998;68:439−443.
587. Konig AL, Keller HE, Braun RW, Roelcke D: Cold agglutinins 608. Ibe M, Rude B, Gerken G, Meyer zum Buschenfelde KH, Lohse
of anti-Pr specificity in rubella embryopathy. Ann Hematol AW: Coombs-negative severe hemolysis associated with hepa-
1992;64:277−280. titis A. Z Gastroenterol 1997;35:567−569.
588. Geisen HP, Roelcke D, Rehn K, Konrad G: High titer cold 609. Melendez HV, Rela M, Baker AJ, et al: Liver transplant for
agglutinins with anti-pr specificity after rubella infection. Klin giant cell hepatitis with autoimmune haemolytic anaemia.
Wochenschr 1975;53:767−772. Arch Dis Child 1997;77:249−251.
589. Roelcke D, Ebert W, Geisen HP: Anti-Pr3: serological and 610. Perez-Atayde AR, Sirlin SM, Jonas M: Coombs-positive
immunochemical identification of a new anti-Pr subspecificity. autoimmune hemolytic anemia and postinfantile giant cell
Vox Sang 1976;30:122−133. hepatitis in children. Pediatr Pathol 1994;14:69−77.
590. Habibi B, Cregut R, Brossard Y, Veron P, Salmon C: Auto-anti- 611. Weinstein T, Valderrama E, Pettei M, Levine J: Early steroid
Pra: A “second” example in a newborn. Br J Haematol therapy for the treatment of giant cell hepatitis with autoim-
1975;30:499−505. mune hemolytic anemia. J Pediatr Gastroenterol Nutr
591. Roelcke D, Kreft H: Characterization of various anti-Pr cold 1993;17:313−316.
agglutinins. Transfusion 1984;24:210−213. 612. Brichard B, Sokal E, Gosseye S, Buts JP, Gadisseux JF, Cornu G:
592. Brody M, Kreysel HW: Cold agglutinin syndrome after rubella Coombs-positive giant cell hepatitis of infancy: Effect of steroids
infection. Kinderarztl Prax 1992;60:134−136. and azathioprine therapy. Eur J Pediatr 1991;150:314−317.
593. Torok TJ: Unusual clinical manifestations reported in 613. Bernard O, Hadchouel M, Scotto J, Odievre M, Alagille D:
patients with parvovirus B19 infection. In: Anderson LJ, Severe giant cell hepatitis with autoimmune hemolytic anemia
Young NS (eds): Human Parvovirus B19. Basel: Karger, in early childhood. J Pediatr 1981;99:704−711.
1997:61−92. 614. Hartman C, Berkowitz D, Brik R, Arad A, Elhasid R, Shamir R:
594. Brown KE: Human parvovirus B19 epidemiology and clinical Giant cell hepatitis with autoimmune hemolytic anemia and
manifestations. In: Anderson LJ, Young NS (eds): Human hemophagocytosis. J Pediatr Gastroenterol Nutr 2001;32:
Parvovirus B19. Basel: Karger, 1997:42−60. 330−334.
595. Kunimi M, Ishikawa K, Tsutsumi H, Hirai M, Kumakawa T, 615. Hadzic N, Portmann B, Lewis I, Mieli-Vergani G: Coombs pos-
Mori M: Parvovirus infection and hemolytic anemia. Am J itive giant cell hepatitis⎯a new feature of Evans’ syndrome.
Hematol 1994;46:159−160. Arch Dis Child 1998;78:397−398.
596. Smith MA, Shah NS, Lobel JS: Parvovirus B19 infection associ- 616. Gurudu SR, Mittal SK, Shaber M, Gamboa E, Michael S, Sigal
ated with reticulocytopenia and chronic autoimmune hemolytic LH: Autoimmune hepatitis associated with autoimmune
anemia. Am J Pediatr Hematol Oncol 1989;11:167−169. hemolytic anemia and anticardiolipin antibody syndrome. Dig
596a.de la Rubia J, Moscardo F, Arriaga F, Monteagudo E, Carreras C, Dis Sci 2000;45:1878−1880.
Marty ML: Acute parvovirus B19 infection as a cause of autoim- 617. Hume R, Williamson JM, Whitelaw JW: Red cell survival in
mune hemolytic anemia. Haematologica 2000;85:995–997. biliary cirrhosis. J Clin Pathol 1970;23:397−401.
597. Petz LD: Hematologic aspects of liver disease. Curr Op 618. Shichiri M, Koyama W, Tozuka S, Sakamoto S, Kanayama M:
Gastroenterol 1989;5:372−377. Primary biliary cirrhosis. A patient with adverse reactions to
597a.Ramos-Casals M, Garcia-Carrasco M, Lopez-Medrano F, et al: tiopronin and autoimmune hemolytic anemia with reticulocy-
Severe autoimmune cytopenias in treatment-naive hepatitis C topenia. Arch Intern Med 1984;144:89−91.
128 Immune Hemolytic Anemias

619. Orlin JB, Berkman EM, Matloff DS, Kaplan MM: Primary 642. Neva FA, Sheagren JN, Shulman NR, Canfield CJ: Malaria:
biliary cirrhosis and cold autoimmune hemolytic anemia: Host-defense mechanisms and complications. Ann Intern Med
Effect of partial plasma exchange. Gastroenterology 1970;73:295−306.
1980;78:576−578. 643. Looareesuwan S, Merry AH, Phillips RE, et al: Reduced ery-
620. Buonanno G, Castaldo C, Izzo GN: Cold agglutinin disease throcyte survival following clearance of malarial parasitaemia
and cryoglobulinemia: Report of two cases. Haematologica in Thai patients. Br J Haematol 1987;67:473−478.
1985;70:174−177. 644. Jeje OM, Kelton JG, Blajchman MA: Quantitation of red cell
621. Lachaux A, Bertrand Y, Bouvier R, Dumont C, Pinzaru M, membrane associated immunoglobulin in children with
Hermier M: Intravenous immunoglobulin therapy in an infant Plasmodium falciparum parasitaemia. Br J Haematol
with autoimmune hemolytic anemia associated with necrotic 1983;54:567−572.
hepatitis and peliosis. J Pediatr Gastroenterol Nutr 1996;22: 645. Berzins K, Wahlgren M, Perlmann P: Studies on the specificity
99−102. of anti-erythrocyte antibodies in the serum of patients with
622. Perseghin P, Balduini CL, Piccolo G, et al: Guillain-Barré syn- malaria. Clin Exp Immunol 1983;54:313−318.
drome with autoimmune hemolytic anemia following acute 646. Wahlgren M, Berzins K, Perlmann P, Bjorkman A:
viral hepatitis. Ital J Neurol Sci 1985;6:447−450. Characterization of the humoral immune response in
623. Yoshioka K, Miyata H: Autoimmune haemolytic anaemia in an Plasmodium falciparum malaria. I: Estimation of antibodies to
asymptomatic carrier of hepatitis B virus. Arch Dis Child P: Falciparum or human erythrocytes by means of
1980;55:233−234. microELISA. Clin Exp Immunol 1983;54:127−134.
624. Tibble JA, Ireland A, Duncan JR: Acute auto immune 647. Lundgren K, Wahlgren M, Troye-Blomberg M, Berzins K,
haemolytic anaemia secondary to hepatitis A infection. Clin Perlmann H, Perlmann P: Monoclonal anti-parasite and anti-
Lab Haematol 1997;19:73−75. RBC antibodies produced by stable EBV-transformed B cell
625. Williams AJ, Marsh J, Stableforth DE: Cryptogenic fibrosing lines from malaria patients. J Immunol 1983;131:2000−2003.
alveolitis, chronic active hepatitis and autoimmune haemolytic 648. Topley E, Knight R, Woodruff AW: The direct antiglobulin test
anaemia in the same patient. Br J Dis Chest 1985;79:200−203. and immunoconglutinin titres in patients with malaria. Trans
626. Pounder RE: Pancreatic exocrine deficiency, chronic active R Soc Trop Med Hyg 1973;67:51−54.
hepatitis and familial autoallergic haemolytic anaemia. Proc R 649. Rosenberg EB, Strickland GT, Yang SL, Whalen GE: IgM anti-
Soc Med 1974;67:323−324. bodies to red cells and autoimmune anemia in patients with
627. Panush RS, Wilkinson LS, Fagin RR: Chronic active hepatitis malaria. Am J Trop Med Hyg 1973;22:146−152.
associated with eosinophilia and Coombs’-positive hemolytic 650. Wenisch C, Spitzauer S, Florris-Linau K, et al: Complement
anemia. Gastroenterology 1973;64:1015−1019. activation in severe Plasmodium falciparum malaria. Clin
628. Lightwood AM, Scott GL: Autoimmune haemolytic anaemia Immunol Immunopathol 1997;85:166−171.
due to red cell antibodies of different specificities in a patient 651. Lefrancois G, Bouvet E, Le Bras J, Vroklans M, Simonneau M,
with chronic hepatitis. Vox Sang 1973;24:331−336. Vachon F: Anti-erythrocyte autoimmunisation during chronic
629. Pengelly CD, Jennings RC: Active chronic hepatitis and falciparum malaria. Lancet 1981;2:661−664.
haemolytic anaemia associated with Rh-specific antibodies. 652. Ritter K, Kuhlencord A, Thomssen R, Bommer W: Prolonged
Postgrad Med J 1971;47:683−686. haemolytic anaemia in malaria and autoantibodies against
630. Cuesta B, Fernandez J, Pardo J, Paramo JA, Gomez C, Rocha E: triosephosphate isomerase. Lancet 1993;342:1333−1334.
Evan’s syndrome, chronic active hepatitis and focal glomeru- 653. Conrad ME: Pathophysiology of malaria. Hematologic obser-
lonephritis in IgA deficiency. Acta Haematol 1986;75:1−5. vations in human and animal studies. Ann Intern Med
631. Christophers SR, Bentley C: Phagocytosis of red cells in black- 1969;70:134−141.
water fever. JAMA 1908;50:1383. 654. Merry AH, Looareesuwan S, Phillips RE, et al: Evidence
632. Zuckerman A: Autoimmunization and other types of indirect against immune haemolysis in falciparum malaria in
damage to host cells as factors in certain protozoan diseases. Thailand. Br J Haematol 1986;64:187−194.
Experimental Parasitology 1964;15:138−183. 655. Petz LD: The expanding boundaries of transfusion medicine.
633. Daniel-Ribeiro CT, Zanini G: Autoimmunity and malaria: In Nance SJ (ed): Clinical and Basic Science Aspects of
What are they doing together? Acta Trop 2000;76:205−221. Immunohematology. Arlington, VA: American Association of
634. Woodruff AW, Ansdell VE, Pettitt LE: Cause of anaemia in Blood Banks, 1991:73−113.
malaria. Lancet 1979;1:1055−1057. 656. Gajewski JL, Petz LD, Calhoun L, et al: Hemolysis of trans-
635. Facer CA, Bray RS, Brown J: Direct Coombs antiglobulin reac- fused group O red blood cells in minor ABO-incompatible
tions in Gambian children with Plasmodium falciparum unrelated-donor bone marrow transplants in patients receiv-
malaria. I: Incidence and class specificity. Clin Exp Immunol ing cyclosporine without posttransplant methotrexate. Blood
1979;35:119−127. 1992;79:3076−3085.
636. Facer CA: Direct Coombs antiglobulin reactions in Gambian 657. Petz LD, Calhoun L, Shulman IA, Johnson C, Herron RM: The
children with Plasmodium falciparum malaria. II: Specificity sickle cell hemolytic transfusion reaction syndrome.
of erythrocyte-bound IgG. Clin Exp Immunol 1980;39:279−288. Transfusion 1997;37:382−392.
637. Facer CA: Direct antiglobulin reactions in Gambian children 658. Kokkini G, Vrionis G, Liosis G, Papaefstathiou J: Cold agglu-
with P: Falciparum malaria. III: Expression of IgG subclass tinin syndrome and hemophagocytosis in systemic leishmani-
determinants and genetic markers and association with asis. Scand J Haematol 1984;32:441−445.
anaemia. Clin Exp Immunol 1980;41:81−90. 659. Pontes De Carvalho LC, Badaro R, Carvalho EM, et al: Nature
638. Abdalla S, Weatherall DJ, Wickramasinghe SN, Hughes M: The and incidence of erythrocyte-bound IgG and some aspects of
anaemia of P: Falciparum malaria. Br J Haematol 1980;46: the physiopathogenesis of anaemia in American visceral leish-
171−183. maniasis. Clin Exp Immunol 1986;64:495−502.
639. Drouin J, Rock G, Jolly EE: Plasmodium falciparum malaria 660. Kobayakawa T, Louis J, Izui S, Lambert PH: Autoimmune
mimicking autoimmune hemolytic anemia during pregnancy. response to DNA, red blood cells, and thymocyte antigens in
Can Med Assoc J 1985;132:265−267. association with polyclonal antibody synthesis during exper-
640. Brown KN, Berzins K, Jarra W, Schetters T: Immune responses imental African trypanosomiasis. J Immunol 1979;122:
to erythrocytic malaria. Clinics in Immunology and Allergy 296−301.
1986;6:227. 661. Wolf CFW, Resnick G, Marsh WL, Benach J, Habicht G:
641. Lee SH, Looareesuwan S, Wattanagoon Y, et al: Antibody- Autoimmunity to red blood cells in babesiosis. Transfusion
dependent red cell removal during P: Falciparum malaria: 1982;22:538−539.
The clearance of red cells sensitized with an IgG anti-D. Br J 662. Homer MJ, Aguilar-Delfin I, Telford SR, III, Krause PJ, Persing
Haematol 1989;73:396−402. DH: Babesiosis. Clin Microbiol Rev 2000;13:451−469.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 129

663. Alward W, Javaid M, Garner J: Babesiosis in a Connecticut thrombocytopenia during a 10 year observation period. Scand
resident. Conn Med 1990;54:425−427. J Haematol 1982;29:265−270.
664. Bowsher B, Callahan CW, Person DA, Ruess L: Unilateral lep- 688. Cunningham-Rundles C, Bodian C: Common variable immun-
tospiral pneumonia and cold agglutinin disease. CHEST odeficiency: Clinical and immunological features of 248
1999;116:830−832. patients. Clin Immunol 1999;92:34−48.
665. Van Audenhove A, Verhoef G, Peetermans WE, Boogaerts M, 689. Oyefara BI, Kim HC, Danziger RN, Carroll M, Greene JM,
Vandenberghe P: Autoimmune haemolytic anaemia triggered Douglas SD: Autoimmune hemolytic anemia in chronic muco-
by Bartonella henselae infection: A case report. Br J Haematol cutaneous candidiasis. Clin Diagn Lab Immunol 1994;1:38−43.
2001;115:924−925. 690. Ballow M, Dupont B, Good RA: Autoimmune hemolytic
666. Girelli G, Teggi A, Perrone MP, Di Vico B, Gandolfo GM, De anemia in Wiskott-Aldrich syndrome during treatment with
Rosa F: Anti-erythrocyte autoimmunization in hydatid transfer factor. J Pediatr 1973;83:772−780.
disease. Int J Clin Lab Res 1993;23:113−115. 691. Cummins L, Spearer W, Stevenson S, et al: Wiskott-Aldrich
667. Issitt PD, Tegoli J, Jackson V, Sanders CV, Allen FH: Anti-IP: syndrome: Clinical, immunologic, and pathologic observa-
Antibodies that show an association between the I and P blood tions. Am J Dis Child 1959;98:579.
group systems. Vox Sang 1968;14:1. 692. Wolff JA: Wiskott-Aldrich syndrome: Clinical, immunologic,
668. Tegoli J, Harris JP, Issitt PD, Sanders CW: Anti-IB, an expected and pathologic observations. J Pediatr 1967;70:221−232.
“new” antibody detecting a joint product of the I and B genes. 693. Douglas SD, Fudenberg HH: Graft versus host reaction in
Vox Sang 1967;13:144−157. Wiskott-Aldrich syndrome: Antemortem diagnosis of human
669. Tippet P, Noades J, Sanger R, et al: Further studies of the I GVH in an immunologic deficiency disease. Vox Sang
antigen and antibody. Vox Sang 1960;13:144. 1969;16:172−178.
670. Perricone R, De Carolis C, Grimaldi O, Fontana L: Activation 694. Pinchas-Hamiel O, Mandel M, Engelberg S, Passwell JH:
of human complement by crude and purified antigens from Immune hemolytic anemia, thrombocytopenia and liver
Echinococcus granulosus. Ric Clin Lab 1984;14:65−72. disease in a patient with DiGeorge syndrome. Isr J Med Sci
671. D’Amelio R, Pontesilli O, Palmisano L, et al: Detection and 1994;30:530−532.
partial characterization of circulating immune complexes in 695. Drobyski WR, Potluri J, Sauer D, Gottschall JL: Autoimmune
hydatid disease. J Clin Microbiol 1983;18:1021−1026. hemolytic anemia following T cell-depleted allogeneic bone
672. von Knorring J, Pettersson T: Haemolytic anaemia complicat- marrow transplantation. Bone Marrow Transplant
ing Yersinia enterocolitica infection: Report of a case. Scand J 1996;17:1093−1099.
Haematol 1972;9:149−152. 696. Godder K, Pati AR, Abhyankar SH, et al: De novo chronic
673. King JW, May JS: Cold agglutinin disease in a patient with graft-versus-host disease presenting as hemolytic anemia fol-
Legionnaires’ disease. Arch Intern Med 1980;140:1537−1539. lowing partially mismatched related donor bone marrow
674. Shurin SB, Anderson P, Zollinger J, Rathbun RK: transplant. Bone Marrow Transplant 1997;19:813−817.
Pathophysiology of hemolysis in infections with Hemophilus 697. Canale VC, Smith CH: Chronic lymphadenopathy simulating
influenzae type b. J Clin Invest 1986;77:1340−1348. malignant lymphoma. J Pediatr 1967;70:891−899.
675. Kaplan KM, Oski FA: Anemia with Haemophilus influenzae 698. Stroncek DF, Carter LB, Procter JL, Dale JK, Straus SE: RBC
meningitis. Pediatrics 1980;65:1101−1104. autoantibodies in autoimmune lymphoproliferative syn-
676. Margolis J: Haemolytic anaemia in Haemophilus influenzae drome. Transfusion 2001;41:18−23.
meningitis treated with specific rabbit serum. Med J Aust 699. Carter LB, Procter JL, Dale JK, Straus SE, Cantilena CC:
1955;1:912−920. Description of serologic features in autoimmune lymphopro-
677. Schiavone DJ, Rubbo SD: Anaemia associated with liferative syndrome. Transfusion 2000;40:943−948.
Haemophilus influenzae meningitis. Lancet 1953;ii:745−749. 700. Straus SE, Sneller M, Lenardo MJ, Puck JM, Strober W: An inher-
678. Caceres W: Severe autoimmune hemolytic anemia associated ited disorder of lymphocyte apoptosis: The autoimmune lym-
with pneumococcal bacteremia. Bol Asoc Med P R 1993;85: phoproliferative syndrome. Ann Intern Med 1999;130:591−601.
16−17. 701. Jackson CE, Fischer RE, Hsu AP, et al: Autoimmune lympho-
678a.Buckley RH: Primary immunodeficiency diseases: Dissectors proliferative syndrome with defective Fas: Genotype
of the immune system. Immunol Rev 2002;185:206–219. influences penetrance. Am J Hum Genet 1999;64:1002−1014.
679. Cunningham-Rundles C: Hematologic complications of 702. Wang J, Zheng L, Lobito A, et al: Inherited human Caspase 10
primary immune deficiencies. Blood Rev 2002;16:61−64. mutations underlie defective lymphocyte and dendritic cell
680. Stiehm ER: Immunologic Disorders in Infants & Children, 4th apoptosis in autoimmune lymphoproliferative syndrome type
ed. Philadelphia: WB Saunders, 1996. II. Cell 1999;98:47−58.
681. Arkwright PD, Abinun M, Cant AJ: Autoimmunity in human 703. van der Werff ten Bosch, Otten J, Thielemans K: Autoimmune
primary immunodeficiency diseases. Blood 2002;99: lymphoproliferative syndrome type III, an indefinite disorder.
2694−2702. Leuk Lymphoma 2001;41:501−511.
682. Robbins JB, Skinner RG, Pearson HA: Autoimmune hemolytic 704. Dianzani U, Bragardo M, DiFranco D, et al: Deficiency of the
anemia in a child with congenital x-linked hypogammaglobu- Fas apoptosis pathway without Fas gene mutations in pedi-
linemia. N Engl J Med 1969;280:75−79. atric patients with autoimmunity/Lymphoproliferation. Blood
683. Sandler SG, Zlotnick A: IgA deficiency and autoimmune 1997;89:2871−2879.
hemolytic disease. Arch Intern Med 1976;136:93−94. 705. Heddle NM, Kelton JG, Turchyn KL, Ali MA: Hyper-
684. Horneff G, Seitz RC, Stephan V, Wahn V: Autoimmune gammaglobulinemia can be associated with a positive direct
haemolytic anaemia in a child with MHC class II deficiency. antiglobulin test, a nonreactive eluate, and no evidence of
Arch Dis Child 1994;71:339−342. hemolysis. Transfusion 1988;28:29−33.
685. Horn B, Viele M, Mentzer W, Mogck N, DeSantes K, Cowan M: 706. Sohngen D, Heyll A, Meckenstock G, et al: Cold agglutinin
Autoimmune hemolytic anemia in patients with SCID after T autoimmune hemolytic anemia as a severe complication in
cell-depleted BM and PBSC transplantation. Bone Marrow B-cell acute lymphocytic leukemia. Transfusion 1992;32:790−791.
Transplant 1999;24:1009−1013. 707. Loughran TP, Jr, Kadin ME, Starkebaum G, et al: Leukemia
686. Nowak-Wegrzyn A, King KE, Shirey RS, Chen AR, of large granular lymphocytes: association with clonal
McDonough C, Lederman HM: Fatal warm autoimmune chromosomal abnormalities and autoimmune neutropenia,
hemolytic anemia resulting from IgM autoagglutinins in an thrombocytopenia, and hemolytic anemia. Ann Intern Med
infant with severe combined immunodeficiency. J Pediatr 1985;102:169−175.
Hematol Oncol 2001;23:250−252. 708. Westbrook CA, Golde DW: Autoimmune disease in hairy-cell
687. Hansen OP, Sorensen CH, Astrup L: Evans’ syndrome in IgA leukaemia: clinical syndromes and treatment. Br J Haematol
deficiency: Episodic autoimmune haemolytic anaemia and 1985;61:349−356.
130 Immune Hemolytic Anemias

709. Tamura H, Ogata K, Yokose N, et al: Autoimmune hemolytic 731. de Misa RF, Suarez J, Medina S, Azana JM, Navas G, Ledo A:
anemia in patients with de novo acute myelocytic leukemia. Cutaneous T-cell lymphoma and autoimmune hemolytic
Ann Hematol 1996;72:45−47. anemia. J Am Acad Dermatol 1992;26:1011−1012.
710. Maldonado NI, Haddock J, Perez-Santiago E: Autoimmune 732. Knox JP, Massa MC, Gradini R, Fu TS, Stiff P, Welykyj SE:
hemolytic anemia in chronic granulocytic leukemia. Blood Autoimmune hemolytic anemia in a patient with mycosis fun-
1967;30:518−521. goides. Cutis 1990;45:52−56.
711. Arbaje YM, Beltran G: Chronic myelogenous leukemia com- 733. Evers J, Albert F, Bazar L, Sulica V, Sacher RA: Autoimmune
plicated by autoimmune hemolytic anemia. Am J Med hemolytic anemia presenting in Sezary syndrome. Report of a
1990;88:197−199. case and review of the literature. Acta Haematol 1992;88:46−49.
712. Cwynarski K, Goulding R, Pocock C, et al: Immune haemolytic 734. Morgensztern D, Kharfan-Dabaja MA, Tsai HM, Lian EC:
anaemia following T cell-depleted allogeneic bone marrow Warm-antibody autoimmune hemolytic anemia developing
transplantation for chronic myeloid leukaemia: Association after thrombotic thrombocytopenic purpura. Acta Haematol
with leukaemic relapse and treatment with donor lymphocyte 2002;108:154−156.
infusions. Bone Marrow Transplant 2001;28:581−586. 735. Bowers D: Sjogren’s syndrome: Systemic lupus erythematosus
713. Lindahl J, Kimby E, Bjorkstrand B, Christensson B, Hellstrom- and auto-immune hemolytic anemia. Report of a case with
Lindberg E: High-dose chemotherapy and APSCT as a prolonged survival. Can Med Assoc J 1969;100:1148−1150.
potential cure for relapsing hemolysing AILD. Leuk Res 736. Boling EP, Wen J, Reveille JD, Bias WB, Chused TM, Arnett FC:
2001;25:267−270. Primary Sjogren’s syndrome and autoimmune hemolytic
714. Sallah S, Gagnon GA: Angioimmunoblastic lymphadenopathy anemia in sisters. A family study. Am J Med 1983;74:1066−1071.
with dysproteinemia: Emphasis on pathogenesis and treat- 737. Schattner A, Shtalrid M, Berrebi A: Autoimmune hemolytic
ment. Acta Haematol 1998;99:57−64. anemia preceding Sjogren’s syndrome. J Rheumatol
715. Freter CE, Cossman J: Angioimmunoblastic lymphadenopathy 1983;10:482−484.
with dysproteinemia. Semin Oncol 1993;20:627−635. 738. Feld S, Landau Z, Gefel D, Green L, Resnitzky P: Pernicious
716. Koo CH, Nathwani BN, Winberg CD, Hill LR, Rappaport H: anemia, Hashimoto’s thyroiditis and Sjogren’s in a woman
Atypical lymphoplasmacytic and immunoblastic proliferation with SLE and autoimmune hemolytic anemia. J Rheumatol
in lymph nodes of patients with autoimmune disease 1989;16:258−259.
(autoimmune-disease-associated lymphadenopathy). Medicine 739. Ramakrishna R, Chaudhuri K, Sturgess A, Manoharan A:
(Baltimore) 1984;63:274−290. Haematological manifestations of primary Sjogren’s syn-
717. Steinberg AD, Seldin MF, Jaffe ES, et al: NIH conference. drome: A clinicopathological study. Q J Med 1992;83:547−554.
Angioimmunoblastic lymphadenopathy with dysproteinemia. 740. Kondo H, Sakai S, Sakai Y: Autoimmune haemolytic anaemia,
Ann Intern Med 1988;108:575−584. Sjogren’s syndrome and idiopathic thrombocytopenic purpura
718. Levey IL: Angioimmunoblastic lymphadenopathy: Compre- in a patient with sarcoidosis. Acta Haematol 1993;89:209−212.
hensive review. Cancer Invest 1987;5:633−647. 741. Sanal O, Ersoy F, Metin A, Tezcan I, Berkel AI, Yel L: Selective
719. Oren H, Ucar C, Gulen H, Duman M, Irken G: Autoimmune IgA deficiency with unusual features: Development of
hemolytic anemia occurring with myelodysplastic syndrome: common variable immunodeficiency, Sjogren’s syndrome,
Report of a pediatric case and review of the literature. Ann autoimmune hemolytic anemia and immune thrombocy-
Hematol 2001;80:540−542. topenic purpura. Acta Paediatr Jpn 1995;37:526−529.
720. Celada A, Farquet JJ, Muller AF: Refractory sideroblastic 742. Schattner A, Friedman J, Klepfish A, Berrebi A: Immune
anemia secondary to autoimmune hemolytic anemia. Acta cytopenias as the presenting finding in primary Sjogren’s syn-
Haematol 1977;58:213−216. drome. QJM 2000;93:825−829.
721. Rojas R, Martin C, Roman J, Garcia JM, Marchal T, Torres A: 743. Jaing TH, Lin KL, Chiu CH, Lo WC, Wu PL: Acute dissemi-
Autoimmune haemolytic anaemia presenting 9 years prior to nated encephalomyelitis in autoimmune hemolytic anemia.
Castleman’s disease. Br J Haematol 1994;86:431−432. Pediatr Neurol 2001;24:303−305.
722. Lerza R, Castello G, Truini M, et al: Splenectomy induced com- 744. Thomson JA, Wilson R, Walker ID: The development of thyro-
plete remission in a patient with multicentric Castleman’s toxicosis (Graves’ disease) during immunosuppression for
disease and autoimmune hemolytic anemia. Ann Hematol autoimmune haemolytic anaemia. Acta Endocrinol (Copenh)
1999;78:193−196. 1986;112:531−535.
723. Marsh JH, Colbourn DS, Donovan V, Staszewski H: Systemic 745. Branehog I, Olsson DS, Weinfeld A, Domellof L: Association of
Castleman’s disease in association with Evan’s syndrome and hyperthyroidism with idiopathic thrombocytopenic purpura
vitiligo. Med Pediatr Oncol 1990;18:169−172. and haemolytic anaemia. Acta Med Scand 1969;205:125−131.
724. Brice P, Marolleau JP, D’Agay MF, Epardeau B, Gisselbrecht C: 746. Brown DM, Lowman JT: Thyrotoxicosis occurring in two
Autoimmune hemolytic anemia disclosing Hodgkin’s disease patients on prolonged high doses of steroids. New Eng J Med
associated with Castleman’s disease. Nouv Rev Fr Hematol 1964;270:278−281.
1991;33:273−274. 747. Chee YC, Gill DS, Poh SC: Hyperthyroidism and autoim-
725. Vaiopoulos G, Kyriakou D, Papadaki H, Fessas P, Eliopoulos mune hemolytic anemia—a case report. Med J Malaysia
GD: Multiple myeloma associated with autoimmune 1978;33:154−155.
hemolytic anemia. Haematologica 1994;79:262−264. 748. Klaassen CH, van Dommelen CK, van Unnik JA: Coincidence
726. Pereira A, Mazzara R, Escoda L, Alcorta I, Nomdedeu B, of adrenal atrophy, haemolytic anaemia and hyperthyroidism
Roelcke D: Anti-Sa cold agglutinin of IgA class requiring in a patient followed by sudden death. Acta Med Scand
plasma-exchange therapy as early manifestation of multiple 1966;180:289−293.
myeloma. Ann Hematol 1993;66:315−318. 749. Olanoff LS, Fudenberg HH: Familial autoimmunity: Twenty
727. Pengelly CD, Mondal BK, Barua AR: Haemolytic anaemia in years later. J Clin Lab Immunol 1983;11:105−111.
myelomatosis. Postgrad Med J 1973;49:279−281. 750. Hennemann HH, Kloss A: Autoimmune haemolytic anaemia,
728. Pirofsky B: Autoimmune hemolytic anemia and neoplasia of thrombocytopenia and thyroiditis: An immunopathological
the reticuloendothelium: With a hypothesis concerning etio- triad. Dtsch Med Wochenschr 1978;103:609−612.
logic relationships. Ann Intern Med 1968;68:109−121. 751. Wenz B, Hasen J: Thyroid disease and red-cell autoantibodies.
729. Cassell M, Chaplin H: Studies on anti-eluate sera. II: N Y State J Med 1978;78:1247−1248.
Comparison of a conventional antiglobulin serum with three 752. Bunin NJ, Carey JL, Sullivan DB: Autoimmune hemolytic
anti-eluate sera in the study of 70 patients with acquired anemia in association with Kawasaki disease. Am J Pediatr
hemolytic anemia. Vox Sang 1960;5:43−52. Hematol Oncol 1986;8:351−353.
730. de Misa RF, Megido M: Autoimmune hemolytic anemia and 753. Hillyer CD, Schwenn MR, Fulton DR, Meissner HC, Berkman
cutaneous T cell lymphoma: Review of the literature. Acta EM: Autoimmune hemolytic anemia in Kawasaki disease: A
Haematol 1993;89:168. case report. Transfusion 1990;30:738−740.
Classification and Clinical Characteristics of Autoimmune Hemolytic Anemias 131

754. Shulman ST: Hemolysis in Kawasaki disease. Transfusion of autoimmune hemolytic anemia complicating thalassemia
1991;31:572. major. Acta Haematol 1990;83:65−68.
755. Cataldo F, Violante M, Bellia L, Gueci G, Maltese I, 765. Wanachiwanawin W, Pootrakul P, Fucharoen S, Piankijagum
Albeggiani A: Autoimmune hemolytic anemia in the course A: High-dose intravenous immunoglobulin in the manage-
of Kawasaki disease. A case presentation. Minerva Pediatr ment of immune hemolysis in patients with thalassemic
1991;43:661−664. disease: Factors which determine refractoriness. Southeast
756. Panzarino V, Estrada J, Benson K, Postoway N, Garratty G: Asian J Trop Med Public Health 1991;22:397−401.
Autoimmune hemolytic anemia after Kawasaki disease in a 766. Besbas N, Ozen S, Bakkaloglu A, Gurgey A, Kanra T, Saatci U:
child. Int J Hematol 1993;57:259−263. Plasma exchange in refractory autoimmune anemia in a child
757. Palazzo E, Oberling F, North ML, Lang JM, Mayer S, Waitz R: with systemic vasculitis associated with homozygote beta tha-
Sarcoidosis in a patient with cold haemagglutinin disease. lassemia. Turk J Pediatr 1994;36:337−340.
Acta Haematol 1972;48:331−336. 767. Cianciulli P, Sorrentino F, Morino L, et al: Radiotherapy com-
758. Thadani U, Aber CP, Taylor JJ: Massive splenomegaly, pancy- bined with erythropoietin for the treatment of extramedullary
topenia and haemolytic anaemia in sarcoidosis. Acta hematopoiesis in an alloimmunized patient with thalassemia
Haematol 1975;53:230−240. intermedia. Ann Hematol 1996;72:379−381.
759. Semple P: Thrombocytopenia, haemolytic anaemia, and sar- 768. Radin AI, Buckley P, Duffy TP: Interferon therapy for agno-
coidosis. Br Med J 1975;5994:440−441. genic myeloid metaplasia complicated by immune hemolytic
760. Andersen E, Skov F, Hippe E: A case of cold haemoglobinuria anemia. Hematol Pathol 1991;5:83−88.
with later sarcoidosis. Treatment with plasmapheresis and 769. Khumbanonda M, Horowitz HI, Eyster ME: Coombs’ positive
immunosuppressiva [sic]. Scand J Haematol 1980;24:47−50. hemolytic anemia in myelofibrosis with myeloid metaplasia.
761. Desablens B, Pruna A, Gontier MF, Messerschmitt J: Auto- Am J Med Sci 1969;258:89−93.
immune haemolytic anaemia associated with bone marrow 770. Ritzmann SE, Levin WC: Cold agglutinin disease: A type of
sarcoidosis. Acta Haematol 1984;71:204−206. primary macroglobulinemia: A new concept. Tex Rep Biol Med
762. Cividalli G, Sandler SG, Yatziv S, Engelhard D, Rachmilewitz 1962;20:236.
N, Rachmilewitz EA: Beta 0-Thalassemia complicated 771. Johnson SA, Oscier DG, Leblond V: Waldenstrom’s macro-
by autoimmune hemolytic anemia. Globin synthesis globulinaemia. Blood Reviews 2002;16:175−184.
during immunosuppressive therapy. Acta Haematol 772. Rosse WF: Clinical Immunohematology: Basic Concepts
1980;63:37−43. and Clinical Applications. Boston: Blackwell Scientific, 1990.
763. Kruatrachue M, Sirisinha S, Pacharee P, Chandarayingyong D, 773. Kyrtsonis MC, Vassilakopoulos TP, Angelopoulou MK, et al:
Wasi P: An association between thalassaemia and autoimmune Waldenström’s macroglobulinemia: Clinical course and
haemolytic anaemia (AIHA). Scand J Haematol 1980;25: prognostic factors in 60 patients. Experience from a single
259−263. hematology unit. Ann Hematol 2001;80:722−727.
764. Argiolu F, Diana G, Arnone M, Batzella MG, Piras P, Cao A: 774. Rytting M, Worth L, Jaffe N: Hemolytic disorders associated
High-dose intravenous immunoglobulin in the management with cancer. Hematol Oncol Clin North Am 1996;10:365−376.
C H A P T E R 4

Mechanisms of Immune
Hemolysis

It has been clear for most of knowledge of phagocytic mononuclear cells did not
this century that two distinct support the use of the terms reticuloendothelial or
types of hemolysis seem to reticulohistiocyte systems.4 They suggested using the
exist. Fairley1 used the term term mononuclear phagocyte system.4 Although the
intravascular hemolysis to concepts are sound, this suggestion has not become
describe the type associated the standard; the term reticuloendothelial system is
with breakdown of red blood still in common usage.
cells (RBCs) directly in the
blood stream, resulting in INTRAVASCULAR IMMUNE
hemoglobinemia and some-
times hemoglobinuria. The term extravascular was RED CELL DESTRUCTION
used when RBCs were destroyed within the reticu-
loendothelial system (RES) without obvious signs of The authors use the term intravascular hemolysis to
hemoglobinemia or hemoglobinuria but rather, an describe complement-mediated RBC destruction in the
increase in the later breakdown products, such as blood stream, in contrast to destruction within the
bilirubin. Dacie2 suggested that a sharp distinction RES. Not many antibodies destroy RBCs intravascu-
does not always hold, as signs of intravascular hemo- larly by a complement-mediated mechanism. Anti-A
lysis (e.g., small amounts of hemoglobin in the plasma) and anti-B are the only examples of alloantibodies that
can be demonstrated in most, if not all, hemolytic destroy RBCs commonly this way; others, such as
anemias if sensitive enough tests are performed (e.g., anti-Jka, -Jkb, -Vel, -PP1Pk, and anti-Lea are capable of
haptoglobin determinations). The authors believe it is causing intravascular lysis of donor RBCs on rare occa-
still convenient to use the terms intravascular and sions, but few others are capable of activating comple-
extravascular as a basis for discussing immune RBC ment efficiently enough to cause formation of the final
destruction, and where the two may overlap is dis- membrane attack complex (see later section).5-7
cussed later in this chapter. Intravascular hemolysis is also uncommon in autoim-
The use of the term reticuloendothelial system has mune hemolytic anemia (AIHA). When it occurs, it is
also been criticized. It was first used by Aschoff in usually associated with paroxysmal cold hemoglobin-
1924.3 The original description of the system of phago- uria (PCH), less commonly with cold agglutinin syn-
cytic cells includes reticular cells of the spleen and drome (CAS) and with drug-induced hemolytic
lymph nodes; reticuloendothelial cells of the lymph anemia secondary to the so-called immune complex
and blood sinuses, including Kupffer cells; histocytes; mechanism (see Chapter 8). Intravascular lysis some-
splenocytes; and monocytes. Criticisms have been times occurs in warm autoimmune hemolytic anemia
focused mainly on the lineage and function of reticu- (WAIHA), more commonly in children than in adults.
lar cells versus histocytes. An international group of Complement-mediated immune hemolysis usually
experts who met in 1969 suggested that the present occurs through the classical pathway of complement

133
134 Immune Hemolytic Anemias

activation. Theoretically, any of the many substances not participate in the alternative pathway of comple-
that can activate the alternative pathway could cause ment activation, but at least three other components—
intravascular hemolysis, but there is, at present, no factors B, D, and P (properdin)—are necessary. In both
evidence to suggest that immune (i.e., antibody- pathways, a series of inhibitors and inactivators
mediated) RBC lysis occurs through this pathway. operate, for example, C1 inhibitor (C1INH) of the clas-
sical pathway and factors H and I, which operate in
Complement Activation both pathways.
Activation of the classical pathway can be initiated
Immunohematologists have been interested in comple- by a number of substances. In immunohematology, it is
ment since the turn of the century when it was first usually immunoglobulin that is the activating agent.
described as a factor present in serum and necessary for Only one molecule of immunoglobulin M (IgM) on the
the lysis of RBCs. Since then, investigators have found cell membrane is necessary to activate the complement
that complement is composed of about 20 plasma system;10,11 it is thought that two subunits have to
proteins, and there are multiple cellular receptors attach to the membrane before activation occurs. In
involved in complement-mediated processes. Muller- contrast, it is thought that immunoglobulin G (IgG)
Eberhard8 defined complement as a multimolecular, needs to form a “doublet,” that is to say, two IgG
self-assembling biological system that constitutes the molecules have to combine with antigens on the cell
primary humoral mediator of antigen-antibody reac- membrane as close together as 250 to 400 Å before they
tions. Complement participates, together with antibody are able to activate C1.10 Only IgG1, IgG2, and IgG3
and cellular elements, in host defense against infection. subclasses can activate complement. Their relative
Also, because of its potential to initiate inflammation efficiency, from highest to lowest, is IgG3, IgG1, IgG2.
and cell destruction, it may become involved in patho- Interaction of antibody with antigen, as, for example,
genic processes, such as tissue injury. Activation of the on RBC membranes, leads to activation of the comple-
complement system may result in (1) membrane ment system, often ending in cytolysis. This involves a
damage (e.g., lysis); (2) production of biologically active series of protein-protein interactions resulting in the
fragments of complement molecules (e.g., C3a and C5a generation of a series of cellular intermediates bearing
anaphylotoxins); and (3) membrane-bound comple- bound complement components designated by
ment components that interact with specific receptors numbers (e.g., EAC1, EAC1,4); the abbreviation EA is
on cells (e.g., RBC–bound C3b interacts with the specific commonly used for the erythrocyte-antibody complex.
C3b receptor on macrophages). The activation process usually is achieved by cleavage
Three pathways of complement activation have of the next complement molecule into fragments. The
been described: the classical, the alternative, and the activated products usually have enzymatic properties
mannose-binding lectin pathways. Figure 4-1 shows (shown by a bar over the numbers); thus, the whole
the three pathways using bacteria as the target,9 but a pathway is an enzymatic cascade similar to the coagu-
RBC can be substituted for the classical and alterna- lation cascade (see Fig. 4-1). The system is held in check
tive pathways. Following activation of any of the by the instability of the complexes formed and the nat-
pathways, the biological consequences of complement urally occurring inhibitors and inactivators present in
activation are similar to those described earlier. normal plasma (e.g., factors H and I).
The pathway consists of three operationally defined
functional units: the recognition unit (C1), the activa-
THE CLASSICAL PATHWAY tion unit (C2, C3, C4), and the membrane attack unit
OF COMPLEMENT ACTIVATION (C5, C6, C7, C8, C9).
The Recognition Unit. C1 is a complex composed of
Eleven major complement components have been C1q and a tetramer C1r2 C1s2. C1r and C1s are serine
described that participate in the classical pathway proteases that are present as proenzymes in the unacti-
(Table 4-1). They are designated numerically: C1 (a tri- vated C1 molecule. Calcium ions are important for the
molecular complex with the subcomponents desig- stability and activation of C1. C1q is a collagen-like
nated C1q, C1r, and C1s), C2, C3, C4, C5, C6, C7, C8, protein with binding sites for IgG and IgM. When C1
and C9. The components circulate throughout the collides with an antigen-antibody complex (e.g., EA), it
extracellular compartment as inactive precursors until is bound to the CH2 domain of the Fc fragment of the
they are activated. This activation often involves immunoglobulin molecule through the C1q subunit.
limited proteolytic cleavage with the formation of An autocatalytic event cleaves C1r to an active form
fragments. One fragment often contributes to the (C1r2), which then acts on C1s2 by cleavage of a single
inflammatory process and the other continues the polypeptide chain (Fig. 4-2). Two regulatory proteins
complement sequence. Fragments of complement are involved in this stage of the reaction. C1q inhibitor
components are designated by a lowercase Arabic (C1qIn) prevents the attachment of complement to the
letter, for example, C3a and C3b. In many publica- immunoglobulin molecule. C1 inhibitor (C1In) abro-
tions, activated complement components that have gates the enzymatic activity of activated C1r and C1s.
enzymatic activity are indicated by placing a bar over The Activation Unit. This unit is assembled in two
the numbered components (e.g., enzymatically active steps (Fig. 4-3). Activated C1 acts on native C4 by
C1 = C1; C3 convertase = C4b,2a). C1, C2, and C4 do cleaving the molecule into C4a and C4b. The major
Mechanisms of Immune Hemolysis 135

FIGURE 4-1. The three activation pathways of complement: the


classical, the alternative, and the mannose-binding pathway. (From
Walport MJ: Complement. N Engl J Med 2001;344:1058–1066.
Copyright © 2001 Massachusetts Medical Society. All rights reserved.)
136 Immune Hemolytic Anemias

TABLE 4-1. PROTEINS PARTICIPATING IN THE C2b


CLASSICAL PATHWAY OF COMPLEMENT ACTIVATION

Serum
Molecular Number Electrophoretic Concentration C1s C4a C2a
Protein Weight of Chains Mobility (mg/mL)

C1q 400,000 18 γ2 180


C1r 190,000 2 β1 100 C4b
C1s 85,000 1 α 80
C2 115,000 1 β1 25
C3 180,000 2 β2 1500
C4 206,000 3 β1 450 C1q
C5 180,000 2 β1 75
C6 128,000 1 β2 60
C7 121,000 1 β2 60
C8 154,000 3 γ1 80
C9 80,000 1 α 150

C2a
fragment C4b binds to the cell membrane. If it remains
C4b
on the cell, it will be cleaved to leave only a small frag-
ment, C4d, on the membrane. A shower of fragments
is produced by a single C1s enzyme so that many C4b
molecules may cluster around the EAC1 site on the _
FIGURE 4-3. If inhibition is overcome, then activated C1 (C1) is capable
cell. The binding site on C4b is short lived and rapidly of cleaving the C4 molecule into two fragments; C4a, which does _ not
decays; if the C4b molecule does not attach to the attach to cells, and C4b, which can attach to cell membrane. C1 also
membrane quickly, it loses its ability to do so. cleaves C2 molecules into C2b, which does not combine with
membranes, and C2a, which may combine with cell-bound C4b to form
Activated C1 also cleaves native C2 into two frag- a complex C4b2a. This complex (C3 convertase) is enzymatic, having
ments (C2a and C2b). The larger C2 fragment, C2a, C3 as its natural substrate.
combines with C4b on the cell membrane to form an
active complex C4b2a (C3 convertase) that has enzy-
see page 109 of reference 7 for a discussion of this pro-
matic activity directed against C3. (Unlike the other
blem.) Magnesium ions are necessary for the formation
complement fragments, most publications use C2a for
of the C4b2a complex. C3 is cleaved by the C4b2a
the larger fragment and C2b for the smaller fragment;
complex into two molecules, C3a and C3b (Fig. 4-4).
The smaller C3a molecule (10,000 d) does not bind to
the cell membrane, but is released into the fluid phase
as a mediator of inflammation (anaphylotoxin I). The
C1r C3b molecule (175,000 d) binds to the cell membrane
C1s and can also bind to its own activation enzyme,
C4b2a. As the C4b2a complex is an enzyme, it can
react more than once and produce a shower of C3b
fragments each time. However, only the C3b frag-
ments that become bound adjacent to the C4b2a
enzyme are believed to participate in the next reaction
in which C5 is cleaved (see Fig. 4-4). These reactions
C1q are affected by various control proteins: factor I in the
presence of the C3b/C4b membrane-associated recep-
tor CR1, C4b-binding protein (C4bp), and decay-
accelerating factor (DAF, CD55). Cell-bound C3b is
cleaved into iC3b, which is cleaved further, leaving
only a small molecule C3dg on the membrane (see
later section).
Antibody Fc Some of the C3b molecules combine with
C4b2a to form C4b,2a,3b (C5 convertase), which will
cleave the C5 molecule into C5a (anaphyloxotin II)
and C5b, which can attach firmly to the RBC
membrane (Fig. 4-5). This is the last enzymatic reac-
FIGURE 4-2. C1q can interact with the Fc portion of immunoglobulin
molecules (an IgG doublet is illustrated). Once a suitable interaction tion in the pathway. During the enzymatic cascade,
occurs, C1r is activated and finally C1s, which possesses esterase only about 5% to 20% of C2, C3, C4, and C5 molecules
activity; its natural substrates are C4 and C2. become bound to the cell membrane.
Mechanisms of Immune Hemolysis 137

The Membrane Attack Complex (MAC). C5b


appears to bind C6 and C7 by adsorption (see
Fig. 4-5). The resulting trimolecular complex attaches
C3a to the cell membrane and binds C8 and C9 (Fig. 4-6).
Fully assembled, MAC consists of about 20 protein
molecules, including one molecule of C5b, C6, C7, and
C8 and multiple molecules of C9. It has a molecular
C3b weight of about 1.7 million. The end result of the
pathway is lysis of the cell.
Electron microscopy shows that lesions start appear-
ing in the cell membrane at the C8 stage, although the
cell does not lyse until C9 is complexed.12 Mayer13 pro-
C2a posed the so-called “doughnut” hypothesis; a stable
hole is produced by the assembly of a rigid, doughnut-
shaped structure in the lipid bilayer of the cell mem-
brane. The hole forms a channel connecting the inside
of the cell with the extracellular fluid. The outside of
the doughnut could be composed of nonpolar pep-
C4b
C3b tides, that is, protein chains that are hydrophobic; the
C3b interior would need polar peptides so that it could be
hydrophilic. He suggested that C5b, C6, C7, C8, and C9
may be the proteins that form the doughnut or funnel
shape, penetrating the lipid bilayer of the membrane.13
FIGURE 4-4. C3 convertase (C4b2a) cleaves C3 into fragments: C3a The cell then lyses either by direct egress of the hemo-
anaphylotoxin, which circulates in the plasma, and C3b, which can globin or, more commonly, through colloid osmotic
attach to cell membranes. Some C3b molecules will fall close to
C4b2a complexes and will form a new enzyme, C4b2a3b or C5 pressure.13 Water and sodium enter the cell with result-
convertase. ant swelling; the membrane of the swollen cell becomes
permeable to macromolecular substances, including
intracellular proteins and nucleotides. Once intracellu-
lar contents have diffused out, the membrane seals
again and effectively becomes an empty sac. The
cellular remnant is removed from the circulation by
phagocytosis.
C5a
C6
C5b

C9
C8
C7

C5b

C2a
C5b
C5b
C6
C7 C6 C6 C7
C6
C4b
C3b
C9 C9 C8

FIGURE 4-5. C5 convertase (C4b2a3b) cleaves C5 into two


fragments: C5a anaphylotoxin, which circulates in the plasma, and
C5b, which is capable of absorbing C6 and C7. The trimolecular FIGURE 4-6. C5b67 absorbs C8. This complex is inserted into the
complex can attach to cell membranes, initiating the formation of the bilipid layer of the membrane. Finally, C9 is absorbed, forming the final
membrane attack complex (MAC). membrane attack complex (MAC), leading to lysis of the cell.
138 Immune Hemolytic Anemias

Several regulatory proteins that affect MAC ment. In 1903, Flexner and Noguchi demonstrated
function have been described, including C8-binding that cobra venom abolished the hemolytic activity of
protein (C8bp) or homologous destruction factor, serum.22 It was shown later that this factor could be
membrane inhibitor of reactive lysis (MIRL, CD 59), isolated from cobra venom, and that it not only inacti-
and S protein in plasma. vated C3 but potentially activated the complement
system at the C3 reaction step, leading to the forma-
THE ALTERNATIVE PATHWAY tion of C5b-9. Studies on cobra venom led to the
OF COMPLEMENT ACTIVATION description of the C3 activator system, with its various
factors, many of which could be equated with factors
In 1954, Pillemer and his coworkers14 showed that described originally by Pillemer. The C3 activator
properdin, a substance present in normal plasma, system was integrated with the properdin system,
would react with a wide variety of naturally occurring providing a basis for an alternative pathway of com-
polysaccharides and lipopolysaccharides to activate plement activation.22
complement, without the presence of antibody. Several The alternative pathway leads to the formation of
other factors present in normal plasma (e.g., C3, factor C5 convertase and thus the self-assembling C5b-9
A, and factor B) were found to be necessary for the complex, with eventual cell damage, just as in the
reaction. They suggested that this might be an impor- classical pathway.
tant in vivo defense mechanism in an unimmunized Reaction Mechanism of the Alternative Comple-
host. Very few scientists accepted Pillemer’s theories at ment Pathway. Table 4-2 lists some activators of the
that time, particularly when work emerged on the alternative pathway. All investigators agree that the
structure of immunoglobulins and the differences alternative pathway can be activated in vitro without
between different immunoglobulin (Ig) classes in com- immunoglobulin (e.g., by endotoxin, inulin, zymosan),
plement activation were described. Many investigators but the ability of nonaggregated immunoglobulin to
believed that the in vitro complement activation that activate the alternative pathway was controversial for
Pillemer had described as attributable to zymosan years. It now seems clear that antibody can play a role
(a yeast extract) was actually the result of a naturally in activation of the alternative pathway. Most workers
occurring IgM antibody to zymosan, and thus the com- agree that aggregated IgA can activate the pathway,23
plement activation was antibody dependent.15 The but there are differing reports in the literature on the
theory of an antibody-independent complement activa- ability of aggregated IgG to activate the pathway.24
tion mechanism lay fallow for about 10 to 15 years and One has to be cautious of interpreting some of the
then was gradually resurrected.16 data, as preparations of immunoglobulins can be con-
In 1968, Gewurz and coworkers17 showed that taminated with endotoxin, which alone can activate
endotoxin and immune complexes could consume the pathway. Evidence for the role of antibody inde-
C3-C9 without affecting C1, C2, or C4 levels. Schur pendent from its classical pathway function has
and Becker18 demonstrated that the F(ab′)2 fragments accrued from experiments with purified proteins of
of rabbit antibody (i.e., rabbit IgG antibody that had the alternative pathway, sera genetically deficient in
the Fc fragment digested away but had both of its C2 and C4, and chelating agents specific for calcium
antigen-combining sites intact) could fix complement. so as to prevent activation of C1. _ _ _
Sandberg and colleagues19 showed that guinea pig Five major proteins (factors P, P, B, B, D) are involved
immune complexes made with IgG1 or IgG2 antibod- solely in alternative pathway activity25 (Table 4-3). Only
ies would fix complement; this was of great interest five of these proteins are essential for initiation and
because guinea pig IgG1 antibodies are known not to amplification because these events occur in the absence
activate C1 (i.e., the antibody does not have a binding of properdin. Of these five proteins, three (C3, factor B,
site for C1 on its Fc fragment). In addition, the IgG1 and factor D) are required for the generation of the
antigen-antibody complexes led to the preferential initial enzyme and the amplifying enzyme of the
fixation of C3-C9 with very little fixation of C1, C4, pathway. The other two proteins (factor H and factor I)
and C2. Ellman and coworkers20 discovered a strain of are regulators of C3 convertase; they suppress C3 con-
guinea pigs that were genetically deficient in C4. This vertase formation in the fluid phase and confine
strain added to the evidence accumulating in support enzyme formation to the surface of activators.
of Pillemer’s original hypothesis, suggesting an alter- Activation of the pathway consists of initiation and
native complement-activation pathway. The F(ab′)2 amplification. Initiation is nonspecific, inasmuch as it
and endotoxin experiments could be duplicated by
the addition of the appropriate antigen-antibody com-
plexes or endotoxins to serum of animals totally TABLE 4-2. ACTIVATORS OF THE ALTERNATIVE
deficient in C4. Bacterial endotoxins and immune COMPLEMENT PATHWAY
complexes depleted the C4-deficient guinea pig sera
of the later complement components, which seemed Immunoglobulins IgA (aggregated)
to prove without doubt that an alternative pathway IgG and IgE (see text)
must be operative.21 Polysaccharides Inulin, agar, endotoxin, yeast,
cell walls
About this time, Muller-Eberhard’s group was Miscellaneous Cobra venom factor, trypsin
studying the interaction of cobra venom and comple-
Mechanisms of Immune Hemolysis 139

TABLE 4-3. PROTEINS PARTICIPATING IN THE ALTERNATIVE PATHWAY

Molecular Electrophoretic
Component Symbol Weight Mobility Synonyms

Properdin P 184,000 γ –
Activated P P– 184,000 γ –
Proactivator B 93,000 β PA, GBG

Activated B B 63,000 γ C3A,GGG,211,Bb

Proactivator Convertase D 24,000 α PAse, GBGase
C3 C3 180,000 β Factor A, HSF
C3b C3b 171,000 α2 HSFa
C3b Inactivator C3b INA 88,000 β
β1H β1H 180,000 β

does not require immunoglobulin or an antibody-like the nonrandom initiation of the classical pathway. A
recognition factor. Initiation appears to be a two-step unique feature of the alternative pathway is a
process involving (1) random binding of C3b through C3b-dependent positive feedback mechanism of
its labile binding site to an activator, and (2) discrimi- amplication. Each newly formed C3b molecule has the
natory interaction of the bound C3b with surrounding potential to form more C3 convertase (C3bBb) and
surface structures. The random event is the result of the thus more C3b.
action of the initial C3 convertase, which is a A discriminatory interaction occurs after binding of
fluid-phase enzyme. C3b to particles (e.g., cells). When bound to a nonacti-
No one has yet described an initiator of the alterna- vator of the alternative pathway, C3b is able to bind
tive pathway that fulfills the role of antibody in the factor H and becomes inactivated through the com-
classical pathway. Lachman and coworkers25,26 sug- bined effects of factors H and I. When bound to an
gested that C3b is formed continuously at a low level activator, control is restricted because C3b is relatively
in a “tick-over” mechanism involving an enzyme. It resistant to inactivation by factors H and I. In contrast,
has also been suggested that the first molecule of binding of factor B to C3b and the formation of C3
metastable C3b may be formed in a nonenzymatic convertase are unaffected by the nature of the surface.
fashion, and that may then be followed by an enzy- As a consequence of the discriminatory phase of
matic step. Native C3 undergoes spontaneous alter- initiation, C3 convertase formation on the activator
ation in neutral buffer at 37°C. Following this occurs, and amplification through the solid phase C3
alteration, a free sulfhydryl group appears that is not convertase commences. Through the additional enzy-
present in native C3. The product, C3(H2O), is a form matic cleavage of C3, more C3b attaches to the
of uncleaved C3 in which the thioester has been membrane; receptor function is thought to reside in at
hydrolyzed. C3(H2O) exhibits all the functional prop- least two critically oriented and closely spaced C3b
erties of C3b, including the ability (unlike native C3) molecules. The role of the second C3b molecule is to
to bind to C3b receptors. C3(H2O) is structurally dis- bind C5 and to modify it for cleavage by Bb. Binding
tinct from C3b in that it contains an intact α-chain, of activated factor B to the C3b-doublet receptor
rather than the α′-chain of C3b that lacks the C3a results in generation of the labile C5 convertase,
domain. Unlike native C3, the α-chain of C3(H2O) is C3b,Bb (which also acts on C3). The C5 convertase
susceptible to cleavage by factor I. Factor I cleaves the can activate properdin (P) nonenzymatically. Pro-
α-chain of C3(H2O) into fragments of 76,000 and perdin is not an essential component for activation
40,000 d. The larger fragment contains the 67,000-d of the pathway, but confers an increased degree of
piece of the α-chain of iC3b plus the covalently linked stability on C3b,Bb, and its presence results in more
9000-d C3a domain of the α-chain. rapid amplification of bound C3b.
In the presence of magnesium, C3b(H2O) and factor Once C3 is activated in the alternative pathway,
B form a reversible complex which, if activated by the the molecular consequences seem to be identical to
serine protease factor D, becomes the initial C3 con- those of the classical pathway. The alternative
vertase C3,Bb. This transient C3 convertase can cleave pathway has not yet been incriminated in many
C3 in the fluid phase into C3a and C3b. Following C3a immunohematologic problems; this may relate to the
removal, C3b expresses a highly reactive site. fact that it is very inefficient in the lysis of human
Metastable C3b can bind covalently to a variety of RBCs as compared to the classical pathway.27 It is of
sugars and attach to cell surfaces. interest to note that the RBCs of patients with PNH
The spontaneous formation of C3(H2O) and thus can be shown to hemolyze through the alternative
the initial C3 convertase provide a mechanism for the pathway as well as the classical pathway.28 It is also
continuous low-level supply of metastable C3b. It has of interest to note that activation of the alternative
been suggested that a random initiation mechanism pathway by autologous RBC stroma has been
operates in the alternative pathway as compared to described.29 Complement activation is controlled by
140 Immune Hemolytic Anemias

TABLE 4-4. PHYSICAL AND FUNCTIONAL PROPERTIES OF THE COMPLEMENT


REGULATORY PROTEINS

MW Serum
State Protein Subunits (kD) (μg/mL) Biological Activity

Soluble C1-INH 1 110 200


——
C4bp 8 500 250 Accelerates decay of C4b2a
——
Factor H 1 150 450 Accelerates decay of C4b2a
Factor I 2 (αβ) 80 35 Cleavage of C3b
S protein 1 83 500 Binds fluid phase C5b-7
Sp-40,40 2 70 50 Binds fluid phase C5b-7
Membrane CR1 1 160–250 Accelerates decay of C3/C5
CD55 1 70 Accelerates decay of C3/C5
MCP 1 45–70 Accelerates decay of C3/C5
HRF/MIP 1 65 Control of MAC formation
CD59 1 20 Control of MAC formation

a number of regulatory proteins; these are shown in excreted in the urine, producing hemoglobinuria.
Table 4-4. During this process, some hemoglobin may be
absorbed by the renal tubules, splitting off iron, which
may eventually (i.e., several days later) appear in the
THE MANNOSE-BINDING LECTIN PATHWAY urine as hemosiderin (also see Chapter 2).
OF COMPLEMENT ACTIVATION Although most blood group antibodies to RBC anti-
gens are IgG1 and/or IgG3, and are thus theoretically
A mannose-binding lectin (MBL), a pattern-recognition
capable of activating C1, most do not activate comple-
molecule of the innate immune system, binds to arrays
ment efficiently enough to cause RBC lysis and/or
of terminal mannose groups on a variety of bacteria.
complement coating of the RBC. Table 4-5 shows the
This reaction can initiate complement activation.30,31
usual complement-activating efficiency of some RBC
MBL interacts with two serine proteases: MSPI, which
alloantibodies. The relative efficiencies probably relate
cleaves C3 directly, and MASP2, which cleaves and
to the number and location of antigen sites on the RBC
activates C3 and C4. This pathway is relevant to bacte-
membrane. For instance, there are at least a million A
ria, but not RBCs.
or B sites on the membrane of A and B RBCs, and the
A and B antigens extend from the membrane, provid-
IN VIVO EFFECTS OF RED CELL–BOUND ing optimal conditions for IgG doublet formation and
COMPLEMENT complement activation. In contrast, there are only
about 30,000 Rh(D) sites, and these are at the mem-
If the complement cascade goes to completion (i.e., brane surface.
MAC formation), then the RBC is hemolyzed. When If the RBC is not hemolyzed, it can circulate as a
hemoglobin is released into the plasma, it rapidly C3b-coated cell. C3b can interact with the macrophage
combines with haptoglobin to form a complex that is CR1 receptor, leading to phagocytosis. Most comple-
cleared within a few hours by the reticuloendothelial ment-coated RBCs are destroyed in the liver by the
system. When the haptoglobin system is saturated, Kupffer cells; they are also destroyed in the spleen,
free hemoglobin circulates in the plasma. Some of it is but the liver has more macrophages than the spleen.
oxidized and bound to albumin as methemalbumin. This is in contrast to the dominant role of the spleen in
Methemalbumin appears about 3 to 6 hours following the destruction of IgG-coated RBCs. The spleen is
a hemolytic episode and remains in the plasma for more efficient than the liver in the destruction of IgG-
about 24 hours. This dark brown pigment may mix coated RBCs because of the reduction of monomeric
with free hemoglobin, leading to brownish-colored plasma IgG (which competes for macrophage Fc
plasma. When plasma contains about 20 mg of hemo- receptors) due to hemoconcentration. As there is no
globin per 100 mL, it will appear slightly pink or light C3b or iC3b in normal plasma, there is no competition
brown, if examined in a thickness of about 1 cm. If for the macrophage complement (CR1 and CR3)
present on the order of 1 mg per mL, the plasma will receptors in the liver; thus the splenic macrophages
appear red.7 Both the hemoglobin-haptoglobin have no more efficiency that the Kupffer cells in the
complex and the methemoglobin are broken down in destruction of C3-coated RBCs. C3b does not remain
the reticuloendothelial system to form bilirubin, which on the red cell very long. If the C3b-coated red cell
will then appear in the plasma (maximum concentra- does not interact, or if the interaction with a C3b
tions are usually not attained for 3 to 6 hours after a receptor on a macrophage is inefficient, the cell-bound
hemolytic episode). When the plasma hemoglobin C3b is denatured. Naturally occurring complement
level exceeds 1 mg per 100 mL, hemoglobin may be control enzymes factors H and I cleave C3b molecules,
Mechanisms of Immune Hemolysis 141

TABLE 4-5. COMPLEMENT-ACTIVATING EFFICIENCY OF RBC


ANTIBODIES

Does Antibody Cause:

In Vitro or In Vivo
In Vivo RBC Complement-Mediated
Complement In Vitro Intravascular
Specificity Sensitization? Hemolysis? Hemolysis?

Anti-A, -B Usually Often Usually


Anti-Rh No No No
Anti-I, -i (Yes)* Rarely† Rarely†
Anti-Lea Usually Sometimes Rarely
Anti-M, -N No No No
Anti-P1 Sometimes Rarely No
Anti-K Sometimes (19%) No No
Anti-Fya Sometimes (13%) No No
Anti-Jka Sometimes (46%) Rarely Rarely
Anti-S Sometimes (33%) No No
Anti-Vel Often Sometimes Sometimes
Anti-PP1Pk Often Sometimes Sometimes

*In vitro sensitization occurs commonly when conditions are optimal (e.g., low temperature, low
ionic strength, or with enzyme-treated RBCs). In vivo sensitization is rare, occurring only when
antibody is of high thermal amplitude (30°C–37°C).
†Only when antibody is of high titer and thermal range.

first forming iC3b, then a second cleavage breaks macrophages32 (Fig. 4-8). They are particularly promi-
away C3c, leaving only C3dg on the RBC membrane. nent in the liver (Kupffer cells), lung (alveolar
C3dg can be further cleaved in vitro, using trypsin, to macrophages), spleen, and bone marrow. The survival
C3d (Fig. 4-7). It should be noted that many earlier time of mature tissue macrophages is thought to be
reports relating to the end in vivo cleavage product several weeks, or even months.32 Immune red cell
use the term C3d when it is now known to be C3dg. destruction occurs predominantly in the spleen and the
C3d is only formed when C3dg is cleaved in vitro liver.7
with enzymes, such as trypsin.
Macrophages have receptors for iC3b (CR1 and
CR3/CD 11b/CD 18) in addition to C3b (CR1/CD35),
Macrophage Receptors
but do not seem to have receptors (or have inefficient Macrophages have receptors on their membranes that
receptors) for C3dg or C3d; thus C3b- or iC3b-coated specifically recognize certain classes of immunoglobu-
RBCs can be destroyed by macrophages within the lins (either monomeric [i.e., not complexed with
reticuloendothelial system. In vivo relationships of antigen] or in an immune complex), and certain com-
complement activation to AIHA are discussed later in plement components. The receptors are for IgG,33-38
this chapter. IgA,39-43 C3,44-46 and C4.45-47
IgG Fc Receptors (FcγR). FcγRs are members of the
EXTRAVASCULAR IMMUNE immunoglobulin superfamily. There are three classes of
Fc receptors: FcγRI, FcγRII, and FcγRIII. Macrophages
RED CELL DESTRUCTION possess all three classes36-38 (Fig. 4-9). The three classes
are encoded by eight genes localized on chromosome 1
If RBCs become sensitized with IgG, or if RBCs are sen- at q21-23. There are at least 12 FcγR isoforms and
sitized with complement but do not proceed through various genetic polymorphisms.36-38
the cascade completely to lysis, then they may be FcγRI (CD64). FcγRI is a 70-kD integral membrane
destroyed or damaged within the RES; this system is glycoprotein expressed predominantly on macrophages
capable of removing up to about 400 mL of RBCs per and monocytes. It binds IgG1 and IgG3 with high
day.7 It is believed that sensitized RBCs are destroyed, affinity, binds IgG4 with a lower affinity, but has little or
within the RES, predominantly by macrophages. no affinity for IgG2. It has a high affinity (kD = 10–8 to
Macrophages arise primarily from bone marrow 10–9 M) for monomeric IgG. This receptor is important
precursors, probably the promonocyte. After a short in the cytoxic lysis of IgG-sensitized RBCs. Interferon
period of maturation in the bone marrow, monocytes regulates FcγRI expression and thus increases antibody-
are released into the blood stream. After spending a few dependent, cell-mediated cytotoxicity (ADCC) effi-
days in the peripheral circulation, they migrate to the ciency. As it is continually reacting with monomeric IgG
tissues and there mature functionally and morphologi- in vivo, FcγRI does not appear to participate in the
cally to become typical histiocytic or exudative initial stages of adherence of IgG-sensitized RBCs to the
142 Immune Hemolytic Anemias

=
S-C
NH2 COOH
S
C3 S S
S

C3 CONVERTASE

C3a R
H O
S C=O
S
S S C3b
S

CR1 / FACTOR H / FACTOR I


FIGURE 4-7. The enzymatic cleavage pathways
of C3. (Reprinted from Freedman J, Semple
R C3f JW: Complement in transfusion medicine. In
H O Garratty G (ed): Immunobiology of Transfusion
S C=O Medicine. New York: Dekker, 1994, by courtesy
of Marcel Dekker Inc.)
S
iC3b S S
S

FACTOR I

R
C3c H O
S C=O C3dg

S
S S
S TRYPSIN

R
H O
C3g S C=O C3d

macrophage. It has been suggested that adherence and IIb1 have a very low affinity for IgG2, but IIaLR
begins with FcγRII and RIII, leading to stripping of the has a similar affinity for IgG2 as for IgG1 (less than for
monomeric IgG from FcγRI and subsequent activation IgG3, but far greater than for IgG4). The IIaLR allotype
of FcγRI to participate in the phagocytic event. is found in only 30% of whites, but is present in almost
FcγRII (CD32). FcγRII is a 40-kD integral membrane 85% of Japanese.33 This polymorphism explains why
glycoprotein that is found on most circulating blood some individuals’ macrophages interact efficiently
cells, except RBCs, in the human circulation. It binds enough with RBCs coated with human IgG2 anti-
all four subclasses, but IgG1 and IgG3 usually bind far bodies to cause AIHA; one would expect to find IgG2-
more efficiently than IgG2 or IgG4. It has a low associated AIHA more commonly in the Japanese.
affinity for monomeric IgG. There is a genetically FcγRII has a low affinity for its ligands, but is impor-
determined polymorphism of the FcγRII receptor tant in phagocytosis, antibody dependent cell medi-
demonstrable by its interaction with mouse mono- ated cytotoxicity (ADCC) superoxide, and lysosomal
clonal IgG1. Three allotypes have been described: enzyme production. FcγRII-mediated ADCC is
IIaHR, IIaLR, and IIb1 (HR = high reactivity with mouse enhanced by IFN-γ. There are three isoforms (RIIA,
IgG1; LR = low reactivity with mouse IgG1). All three RIIB, and RIIC) of FcγRII. RIIA induces phagocytosis;
allotypes have a high affinity for IgG3. Allotypes IIaHR RIIB and RIIC only induce adherence of EA to
Mechanisms of Immune Hemolysis 143

Macrophages
Spleen
Liver
Lung
Lymph Nodes
Stem Cell etc...

Monoblast

Promonocyte

Monocytes
Monocyte

FIGURE 4-8. Mononuclear phagocyte cell lineage. Hematopoietic stem cells in the bone marrow give rise to monoblasts that divide and produce
promonocytes. The promonocytes differentiate into monocytes that leave the bone marrow and circulate in the blood. The monocytes migrate into
tissues where they differentiate into macrophages. (Reprinted from Horsewood P, Kelton JG: Macrophage-mediated cell destruction. In Garratty G
(ed): Immunobiology of Transfusion Medicine. New York: Dekker, 1994:435–464, by courtesy of Marcel Dekker Inc.)

Fcγ RI Fcγ RII Fcγ RIII

GPI

61 aa 30 aa 76 aa 52 aa 44 aa 25 aa
FIGURE 4-9. Schematic diagram of the three classes of membrane Fcγ receptors. The FcγRI integral membrane receptors comprise three
immunoglobulin C2 domains and a crytoplasmic tail of either 61 or 30 amino acids (aa). The FcγRII integral membrane receptors have two extracellular
immunoglobulin domains and a cytoplasmic tail of 76, 52, or 44 amino acids. The integral membrane FcγRIII that is present on macrophages and NK
cells has two immunoglobulin domains and a 25-amino acid cytoplasmic tail. The FcγRIII present on neutrophils is a glycosylphosphatidylinositol
(GPI)–linked molecule lacking a cytoplasmic tail, and it has two extracellular immunoglobulin domains. (Reprinted from Horsewood P, Kelton JG:
Macrophage-mediated cell destruction. In Garratty G (ed): Immunobiology of Transfusion Medicine. New York: Dekker, 1994:435–464, by courtesy of
Marcel Dekker Inc.)

macrophages.38 Indik and coworkers38 found that killer (NK) cells. It binds IgG1 and IgG3, but not IgG2
FcγRIIA consistently mediated higher levels of or IgG4. It has a low affinity for monomeric IgG (kd
phagoctyosis than FcγRI or RIII. <10–7 M). Two FcγRIII genes have been identified. The
FcγRIII (CD16). FcγRIII is a 45- to 65-kD membrane FcRIIIA gene codes for a transmembrane glycoprotein
glycoprotein present on a subpopulation of monocytes, expressed on macrophages and NK cells. The FcRIIIB
macrophages, neutrophils, eosinophils, and natural gene codes for a glycosylphosphatidylinositol (GPI)-
144 Immune Hemolytic Anemias

linked protein expressed on neutrophils only; this Complement Receptor Type 1 (CR1 [CD35]). CR1
protein carries the NA1/NA2 neutrophil-specific is found on most circulating cells, including RBCs. It
antigen. The macrophage FcγRIII can mediate phago- is a large (190 to 280 kD) molecule projecting above
cytosis of IgG-coated RBCs. the glycocalyx of the membrane. The extracellular
IgA Fc Receptors. Although early work suggested part of the molecule is composed of four long homo-
that macrophages did not have an IgA receptor, such logous repeats, each of which is composed of seven
a receptor (FcαR) has now been described by several short consensus repeats. Each of the long repeats con-
groups.39-43 One IgA receptor (Fcα/μR) has recently stitutes a separate receptor site, except for the one
been shown to crossreact with IgM.47a Its clinical nearest the membrane. The repeat furthest from the
significance in RBC destruction is unknown. membrane is the C4b3b receptor. The two more cen-
trally located repeats are receptors for C3b. CR1, on
macrophages, can participate in phagocytosis of sen-
Complement Receptors sitized RBCs, but a second signal (e.g., IgG and Fc
receptor) is usually required. CR1 acts as a receptor
There are three complement receptors on macrophages: for C3b, iC3b, C4b, and C1q. C3b interactions are the
CR1, CR3, and CR445,46 (Fig. 4-10). most efficient. C4b and C1q are very inefficient.

CR 1 CR 3
F (or A) Allotype

LHR A C4b Binding Site CD11b

SCR
CD18
B

C3b Binding Sites A (or I) Domain

HTR

D CRR

FIGURE 4-10. Schematic diagram of the structure of the CR1 and CR3 complement receptors. The CR1 F (also known as A) allotype is shown. It is
a single-chain glycoprotein organized into four (A–D) long homologous repeats (LHRs), each composed of seven short consensus repeats (SCRs).
The terminal LHR binds C4b and the two central LHRs bind C3b. CR3 is a noncovalently linked heterodimer composed of an α chain (CD11b) and a
β chain (CD18). The α chain (CD11b) contains seven homologous tandem repeats (HTR), the second and third of which are split by an additional A
(also called interactive, I) domain that may contain the C3 ligand-binding site. The β chain (CD18) has a cysteine-rich region (CRR) consisting of four
tandem repeats. (Reprinted from Horsewood P, Kelton JG: Macrophage-mediated cell destruction. In Garratty G (ed): Immunobiology of Transfusion
Medicine. New York: Dekker, 1994:435–464, by courtesy of Marcel Dekker Inc.)
Mechanisms of Immune Hemolysis 145

Red cells have relatively few CR1 receptors Complement Receptor Type 4 (CR4). CR4 is closely
(500–700/red cell), but as there are so many RBCs in related to CR3 and also recognizes iC3b. Like CR1 and
the body, they become the major CR1-carrying cell CR3, to participate in phagocytosis, CR4 needs an
population. This suggests a major role for RBCs in additional signal.
clearing complement (C3b, iC3b)-carrying immune
complexes from the circulation. These complexes are
stripped from the RBCs by high-affinity CR3 receptors Macrophage Interactions with
on macrophages in the RES, particularly Kupffer cells RBCs Coated with Immunoglobulin
in the liver. The RBCs appear not to be harmed by this and/or Complement
process and return to the circulation (see later section).
The Knops system blood group antigens are found The attachment of appropriately sensitized RBCs to
on the CR1 molecule.48 macrophages can be easily visualized in vitro. Some-
Complement Receptor Type 3 (CR3 [CDIIb/CD18]). times, there is a so-called rosette formation whereby
CR3 is the receptor for iC3b. It is widely distributed the macrophage becomes ringed by sensitized RBCs,
(e.g., in macrophages, monocytes, follicular dendritic like petals on a flower (Fig. 4-11).49 Upon attachment
cells, neutrophils, and eosinophils). CR3 is a member of to the macrophage, the RBCs usually undergo
the integrin superfamily of adhesion molecules. It can considerable distortion and deformity in the region of
mediate phagocytosis and lysis of iC3b-coated RBCs; the attachments.49 The sensitized RBC may become
like CR1, a second signal is probably required for completely engulfed by the macrophage and may be
phagocytosis. CR3 is also involved in killing of cells by destroyed internally (Fig. 4-12), or a portion of the
NK cells. RBC may become internalized (Figs. 4-13 and 4-14),

FIGURE 4-11. Scanning electron micrograph


illustrating the interaction of antibody-coated
red cells and phagocytic white cell. The white
cell is surrounded by sensitized red cells
forming a rosette (courtesy of Dr. W. Rosse).
146 Immune Hemolytic Anemias

FIGURE 4-12. Scanning electron micrograph illustrating the reaction of


a phagocytic white cell with an antibody-coated red cell. Only a small
exposed area remains of a nearly ingested red cell. (Reproduced from
Rosse WF, de Boisfleury A, and Bessis M: The interaction of phagotic FIGURE 4-14. Further interaction between the phagocytic white cell
cells and red cells modified by immune reactions: Comparison of and the antibody-coated red cell results in internalization of a portion of
antibody and complement coated red cells. Blood Cells 1975;1:345, by red cell. (Reproduced from Rosse WF, de Boisfleury A, and Bessis M:
permission of Springer-Verlag.) The interaction of phagotic cells and red cells modified by immune
reactions: Comparison of antibody and complement coated red cells.
Blood Cells 1975;1:345, by permission of Springer-Verlag.)

FIGURE 4-13. Phase-contrast photomicrograph illustrating the


interaction of an antibody-coated red cell and a phagocytic white cell.
The red cell, having been taken by the metapod, is deformed as the FIGURE 4-15. The separation of the internal and external portions of
metapod flows along its sides. (Reproduced from Rosse WF, de the red cell is complete; the portion of the red cell outside the
Boisfleury A, and Bessis M: The interaction of phagotic cells and red macrophage may escape and circulate as a spherocyte. (Reproduced
cells modified by immune reactions: Comparison of antibody and from Rosse WF, de Boisfleury A, and Bessis M: The interaction of
complement coated red cells. Blood Cells 1975;1:345, by permission phagotic cells and red cells modified by immune reactions: Comparison
of Springer-Verlag.) of antibody and complement coated red cells. Blood Cells 1975;1:345,
by permission of Springer-Verlag.)
resulting in loss of membrane protein and lipids. The
portion of the RBC outside the macrophage may by enzymes released by the macrophage (ADCC).
escape and circulate as a spherocyte50 (Fig. 4-15). The Figure 4-16 summarizes the possible interactions of a
membrane of this spherocytic RBC is rigid owing to sensitized RBC with macrophages.
the loss of protein and lipids; thus the RBC is unable
to change its shape readily to traverse the fine chan- MACROPHAGE/MONOCYTE CYTOTOXICITY
nels of the spleen and so is susceptible to early
destruction.51-53 Once attached to the macrophage, a Since the incrimination of macrophages as the major
sensitized RBC may also be destroyed or damaged effector cell in immune RBC destruction, the emphasis
Mechanisms of Immune Hemolysis 147

FIGURE 4-16. Immune RBC destruction by cellular


mechanisms.

has been on phagocytosis. However, recent studies promoting phagocytosis or lysis per molecule of
have suggested that an extracellular mechanism is IgG bound; however, because of the greater antigen
also operative and that the cytotoxic properties of the density of A and B, more than 100,000 molecules
macrophage may be more important than first of IgG per RBC could be bound, producing equiva-
thought.54 Most of the interactions are similar to those lent lysis of anti-D sensitized cells. Even after degra-
described previously for ADCC by lymphocytes: dation of C3b to C3d, complement augmentation
(1) specific recognition and cell adherence; (2) intimate persisted.
contact stimulating membrane phagocytosis and trig- Engelfriet’s group54,67-70 has suggested that cytotoxic-
gering release of specific macrophage cytotoxin; (3) ity may play a more important role than phagocytosis in
specific macrophage cytotoxin acting directly on the immune RBC destruction. Using 51Cr-labeled human
target cell membrane. RBCs sensitized with anti-D, they were able to demon-
There are many publications showing that mono- strate cytotoxicity by monocytes, independent of
cytes/macrophages can cause lysis (ADCC) of RBCs phagocytosis. An interesting finding was that the cyto-
sensitized with anti-A, anti-B, or anti-D.55-70 For toxicity, but not the phagocytosis, was inhibited by
instance, Kurlander and colleagues56 were able to hydrocortisone.70 The release of lysosomal enzymes by
demonstrate in vitro monocyte-mediated lysis of the monocytes was also inhibited by hydrocortisone; a
human RBCs sensitized with IgG anti-Rh (D) and significant correlation was found between lysosomal
anti-A or -B. Cells sensitized with only human com- enzyme release and lysis.70 The authors concluded that
plement components (even up to 80,000 molecules lysosomal enzymes released by monocytes, when incu-
per RBC) were not lysed, but complement (C3b or bated with Rh-sensitized RBCs, were responsible for
C3d) sensitization augmented IgG-mediated lysis lysis of these RBCs. The lysis occurred only over a short
and reduced the amount of IgG necessary to produce range, probably at the site of attachment of the red cell,
lysis. Without complement, 1000 to 1500 molecules because only RBCs bound to the monocyte were lysed.
of anti-D per RBC were necessary for lysis; less than Thus, in vivo IgG-sensitized RBCs may attach to
1000 molecules were necessary when complement macrophages in the spleen and be lysed external to the
was present in addition to IgG on the RBC. IgG macrophage, without phagocytosis occurring. This
anti-A or anti-B was 5- to 10-fold less efficient in direct lysis (ADCC) would not require complement.
148 Immune Hemolytic Anemias

Unlike other investigators,71-85 Fleer and coworkers67,68


TABLE 4-6. FACTORS THAT INFLUENCE
were not able to demonstrate lysis of Rh-sensitized cells
THE PATHOGENICITY OF RBC ANTIBODIES
by lymphocytes. Another important effect noted by
Fleer and coworkers59 was the development of an
Characteristics of antibody
increased osmotic fragility of a considerable part of the Class
nonlysed Rh-sensitized red cells; osmotically more Subclass
fragile RBCs considerably outnumbered, lysed, and Specificity
ingested RBCs. A correlation with increased osmotic Thermal range
Complement-activating efficiency
fragility and severity of autoimmune hemolytic anemia Affinity
had been described previously.86 The presence of sphe- As yet unknown qualitative characteristics
rocytes has, for years, been said to be a hallmark of Quantity of RBC-bound IgG and/or complement
AIHA; the spherocytes are now thought to be formed Characteristics of target antigen
through fragmentation of the sensitized RBCs by Quantity of antigen on membrane
Distribution of antigen on membrane
macrophages49 (see Figs. 4-13 to 4-15). It has also been Presence of antigen in tissues and/or body fluids
assumed that the increased osmotic fragility is due to Type of complement present on circulating RBCs
the presence of spherocytes; it may well be that the Activity of reticuloendothelial system
damage to the RBC membrane caused by cytotoxicity,
without fragmentation and spherocyte formation, may
also contribute to the increased fragility. organ, it has been shown that the spleen is nearly
Extravascular red cell destruction leads to the 100 times more efficient at removing Rh (IgG)–
appearance, in the plasma and urine, of breakdown sensitized RBCs.7 About 20 to 30 μg of anti-Rh per mil-
products of hemoglobin, such as bilirubin and uro- liliter of RBCs will lead to complete clearance in a
bilinogen.7 Occasionally, laboratory tests may show single passage through the spleen (T50 of 20 minutes),
some results that are associated with intravascular but in the absence of the spleen, the cells are removed
lysis (e.g., hemoglobinemia and hemoglobinuria), by the liver with a T50 of about 5 hours.7 Lo Buglio and
even though no complement-mediated lysis has coworkers49 showed that the reaction between the Fc
occurred. For instance, hemoglobinemia and hemo- receptor on human monocytes and macrophages with
globinuria are relatively common following destruc- IgG-sensitized RBCs could specifically be inhibited
tion of large volumes of Rh-incompatible blood (see with IgG, or its Fc fragment, in solution. They sug-
Chapter 14). The Rh antibodies in these cases have gested that the free IgG in plasma competes with
never been shown to fix complement. As macro- RBC–bound IgG for receptors on the monocyte/
phages (and possibly, lymphocytes) are known to macrophage surface. At hematocrit levels in excess of
destroy sensitized RBCs by extracellular cytotoxicity 75%, comparable to those encountered in the splenic
(ADCC) in addition to phagocytosis, this may explain red pulp, plasma had little inhibitory effect. It was sug-
the noncomplement-mediated hemoglobinemia and gested that plasma skimming enhanced entrapment of
hemoglobinuria associated with extravascular lysis. In antibody-coated RBCs in vivo; the fact that the spleen is
addition, or perhaps alternatively, hemoglobin may be notable for both erythroconcentration and for efficient
released into the blood stream following fragmenta- trapping of IgG-coated RBCs supported this interpreta-
tion of the RBCs during phagocytosis, particularly tion. Other investigators54,87-89 have confirmed the
when large amounts of blood are rapidly destroyed inhibitory effect of even small amounts of normal
(e.g., several units).5 Most alloantibodies (e.g., Rh, Kell, plasma or serum.
Duffy) other than ABO and the few rare antibodies Fleer and coworkers87 published data supporting the
mentioned previously destroy RBCs extrasvascularly. hypothesis of Lo Buglio and coworkers,49 that is, that
Most of the RBC destruction associated with AIHA is the efficiency of splenic destruction was related to the
extravascular (e.g., warm antibody AIHA, which com- hemoconcentration (loss of plasma) that occurs in the
prises almost 70% of all AIHA). In addition, many of spleen. They were able to show that the in vitro inter-
the drug-induced hemolytic anemias (e.g., those action between monocytes and IgG Rh–sensitized
caused by methyldopa or penicillin) are primarily RBCs was completely inhibited by low concentrations
associated with extravascular red cell destruction. of IgG (e.g., 30 to 100 μg/mL); however, the interaction
Many factors may affect immune red cell des- with IgG anti-A–sensitized RBCs was not inhibited by
truction.6 The more important factors are listed in Table IgG. They suggested that this difference was probably
4-6. Some of these factors are discussed relative to: quantitative. The higher the number of IgG antibody
(1) RBCs sensitized with Ig alone, and (2) RBCs sensi- molecules per sensitized RBC, the less the interaction
tized with complement, with or without Ig present. between the sensitized RBC and monocyte was inhib-
ited by IgG. Kelton and coworkers88 showed that there
was a significant relationship between the concentra-
DESTRUCTION OF RED CELLS SENSITIZED tion of IgG in the serum and the rate of clearance of
WITH IgG ALONE antibody-sensitized RBCs (r = 0.51, p < 0.1). Using
51Cr-labeled IgG (Rh)–sensitized RBCs, they showed
RBCs sensitized by IgG only are destroyed predomi- that patients with hypergammaglobulinemia had the
nantly in the spleen. Although the liver is a much larger slowest Fc-dependent clearance, whereas those with
Mechanisms of Immune Hemolysis 149

hypogammaglobulinemia had the most rapid clear- also reported that the amount of phagocytosis was
ance. The unusually rapid clearance in a patient with greatly influenced by the proportion of sensitized
hypogammaglobulinemia could be returned to normal RBCs, when in the presence of plasma IgG.
by raising the concentration of IgG in the serum. This Quantitative Factors. Generally speaking, there
would seem to support the observations by Lo Buglio appears to be a direct relationship between the amount
and coworkers49 and Fleer and coworkers,87 but does of IgG on RBCs and the amount of in vivo RBC destruc-
not agree with the findings reported by Scornik and tion. This is generally true for alloantibodies, but there
coworkers.90 The latter showed that IgG-sensitized are many exceptions to this when autoantibodies are
RBCs were rapidly cleared from the circulation of studied. Mollison7 has clearly shown that if different
patients with myeloma, despite serum IgG concentra- quantities of a single example of an allo anti-Rh are
tions that were several times higher than normal. They used to sensitize RBCs in vitro, there is a good correla-
concluded that the current explanations derived from tion between the amount of IgG sensitizing the red cell
in vitro experiments were insufficient to explain the and the rate of in vivo clearance (Fig. 4-17).
IgG-dependent clearance of RBCs in the presence of Constantoulakis and colleagues91 also showed this, but
free IgG. in studying autoantibody sensitization, they were not
Fleer and coworkers87 reported that the number of able to establish a close correlation between the amount
sensitized RBCs per monocyte also had a strong of antibody on the RBC and the rate of destruction
influence on the inhibitory effect of IgG. When the determined by 51Cr. In contrast to this, Rosse92 found
number of sensitized RBCs per monocyte was that the amount of RBC–bound IgG was generally pro-
increased from 1 to 32, the percentage of inhibition by portional to the rate of RBC destruction, although
a fixed amount of IgG (50 μg/mL) decreased many exceptions could be found. In a given patient, the
significantly. This in vitro effect was only evident rate of destruction was closely related to the concentra-
when relatively weakly sensitized RBCs were used tion of IgG antibody detected on the membrane; this is
and when the in vivo destruction of these weakly sen- of some value in follow-up evaluation of an individual
sitized RBCs was confined to the spleen. As a consid- patient during response to therapy (see Chapter 11). It
erable hemoconcentration occurs in this organ, it is has been known for some time that RBCs sensitized
conceivable that a highly sensitized RBC to with the same amounts of IgG autoantibody can have
macrophage ratio is accomplished. A high ratio may markedly different cell survival times. For example,
allow interaction between weakly sensitized RBCs Constantoulakis and coworkers91 reported two differ-
and splenic macrophages, despite the presence in vivo ent RBC samples with approximately 0.9 μg of IgG
of a high concentration of IgG. Kurlander and Rosse89 antibody nitrogen on each having a T50 of 45 minutes

100 1• 6

80
4• 2
60
6• 2 9• 3

40
FIGURE 4-17. Survival of Rh-positive RBCs sensitized 8• 2
in vitro with varying amounts of a particular anti-Rh
Percentage survival

(Avg). Purified IgG, prepared from Avg serum, was 13• 4


used to sensitize the RBCs. The figures against each
slope are the estimates of the amount of antibody on 28• 4 29• 0
the RBCs at the time of injection (expressed as 32• 0
grams of antibody per milliliter red cells). In three 20
cases (x) the estimates were made by using labeled
anti-Rh. (From Mollison PL, Crome P, Hughes-Jones
NC, Rochna E: Rate of removal from the circulation
of red cells sensitized with different amounts of
antibody. Brit J Haematol 1965;11:401.)
10 47• 0

43• 3
41• 5
5
0 20 40 60 80
Minutes
150 Immune Hemolytic Anemias

and 11 days, respectively. Most patients who have a Garratty and Nance99 used flow cytometry to study
positive DAT result due to methyldopa have normal 103 patients, with and without hemolytic anemia, with
RBC survival, although these RBCs can be extremely positive DAT results. Ten of the positive DATs were
strongly sensitized with IgG (see Chapter 8). Similarly, determined to be idiopathic (seven were associated
a small proportion of normal, healthy blood donors have with hemolytic anemia and three were not). Twenty-
a strongly positive DAT result, with normal RBC sur- five positive DAT results were methyldopa-
vival. It is still not understood why this is so.6,93-96 When induced (eight were associated with hemolytic anemia
macrophages were found to have IgG1 and IgG3 and seventeen were not). Thirty-eight of the patients
receptors but not IgG2 or IgG4 receptors, it was were neonates with positive DATs born to mothers
thought that this might explain the discrepancies. who had IgG alloantibodies of potential clinical
Although some rare cases can be explained by the significance (13 of these babies had hemolytic anemia
subclass of IgG present on the RBC (e.g., IgG4 does not and 25 did not). Thirty were normal donors with posi-
activate complement or have a receptor site for tive DATs and no evidence of hemolytic anemia, and
macrophages), this is not usually the answer (see later 121 were normal donors with negative DAT results.
section). One of the problems with relating the quantity The amount of RBC–bound IgG was compared using a
of RBC–bound IgG and the degree of in vivo destruc- fluorescein-conjugated anti-IgG. Preliminary experi-
tion has been the difficulty in accurately measuring the ments showed that flow cytometry was capable of dif-
number of IgG molecules per RBC when the RBCs were ferentiating the amounts of red cell–bound IgG on
strongly sensitized (e.g., in patients with a 4+ DAT). strongly sensitized RBCs, even when serologic assays
Hughes-Jones97 showed that RBCs that were strongly (e.g., antiglobulin test titration scores) could not do so.
sensitized (e.g., in patients with a 4+ DAT) with an IgG Table 4-8 shows the results of testing RBCs sensitized
Rh antibody could have from 30% to (theoretically) in vitro with decreasing amounts of anti-D; all the
100% antigen saturation (Table 4-7). Although the RBCs reacted 4+ by antiglobulin test, and the titration
macrophage may notice these differences, until scores (i.e., from tests using dilutions of antiglobulin
recently, it has been difficult to accurately compare RBC serum) were similar. Flow cytometry (using an Ortho
IgG sensitization in individuals within this range. Spectrum flow cytometer) clearly differentiated the
Using flow cytofluorometry, van der Meulen and degree of sensitization when the amount of fluores-
coworkers98 suggested that it was predominantly the cence (i.e., anti-IgG) per RBC in region B (i.e., those
quantity of RBC–bound IgG autoantibody that deter- RBCs with greater than normal amounts of IgG) was
mined whether or not in vivo hemolysis occurred. measured. Table 4-9 shows the results of measuring
They studied 29 patients who had RBCs sensitized the RBC–bound IgG in the patients and donors
with IgG1 antibodies (7 of them also had C3d on their described earlier. It can be seen that the mean was
cells). Twelve of these patients had hemolytic anemia higher in all groups when hemolytic anemia was
and 17 did not. Fifteen of the 29 patients were receiv- present. However, unlike the study by van der Meulen
ing methyldopa; 9 of these had hemolytic anemia and and colleagues,98 no hemolytic threshold was obvious,
6 did not. The relative amount of RBC–bound IgG was and there was considerable overlap between patients
measured by flow cytometry using a fluorescein- with and without hemolysis in each group.
labeled anti-IgG1. With the exception of one patient Merry and coworkers100,101 were successful in using
with hemolytic anemia and one without hemolysis, 125I-labeled anti-IgG to measure RBC–bound IgG, even
flow cytometry (which measures the amount of when the cells were strongly sensitized. Using this
RBC fluorescence, which, in turn, relates directly to assay, they also found a general correlation between the
the amount of RBC–bound IgG) clearly differentiated amount of RBC–bound IgG and in vivo hemolysis.
the group with hemolytic anemia from those without. However, like Garratty and Nance,99 they could not
In the methyldopa-induced group, there was a determine a quantitative hemolytic threshold. More
perfect correlation. These results suggested that recently, other investigators have published similar
there is a quantitative in vivo hemolytic threshold, results102-104 that have supported the conclusions of
and that flow cytometry can be used to identify this Garratty and Nance.99 Sokol reviewed the literature on
threshold. the quantitation of RBC–bound proteins.105

TABLE 4-7. COMPARISON BETWEEN THE STRENGTH OF AGGLUTINATION OF THE ANTIGLOBULIN TEST
AND THE DEGREE OF SATURATION OF THE ANTIGEN SITES WITH 131I-LABELED ANTI-D97

Dilution of 131I-labeled Anti-D Neat 1:2 1:4 1:8 1:16 1:32 1:64 1:128 1:256 1:512

Strength of agglutination 4+ 4+ 4+ 4+ 4+ 3+ 3+ 2+ 1+ 1⁄2+


Percentage saturation 87 75 61 48 30 17 9 5 2.6 1.8
of antigen sites
Mechanisms of Immune Hemolysis 151

Garratty studied the IgG subclass of RBC–bound


TABLE 4-8. DEMONSTRATING RELATIVE AMOUNTS
IgG of 89 normal blood donors with no evidence of
OF RBC-BOUND IgG BY FLOW CYTOMETRY99
hemolytic anemia and of 78 patients with positive
DAT results, with and without hemolytic anemia.6
Flow Cytometry
(Mean Table 4-10 shows the results. In all three groups, RBCs
Anti-D Indirect Antiglobulin Fluorescence were sensitized with predominantly IgG1 autoanti-
Dilution Test Score per RBC) bodies (89%, 85%, and 87%, respectively); usually,
IgG1 was the only subclass present. IgG2 was present
1 in 40 65 84.7 alone, or together with other subclasses, on 5.8%,
1 in 20 62 130.1
1 in 10 68 183.2 15.6%, and 26.2%, respectively, of the RBCs from
1 in 5 68 212.0 donors and patients without and with hemolytic
anemia. IgG2 was not found often as the only subclass
on the RBCs, but it is interesting to note that one
patient with hemolytic anemia had only IgG2
detected on the RBCs (see later section). IgG3 was
TABLE 4-9. RELATIONSHIP OF THE AMOUNT detected on 7%, 13%, and 29%, respectively, of the
OF RBC-BOUND IgG (MEASURED BY FLOW RBCs; similar to IgG2, it was unusual to find IgG3
CYTOMETRY) AND HEMOLYTIC ANEMIA99 alone on the RBCs. IgG4 was detected on 12%, 13%,
and 24%, respectively, of the RBCs; it was detected as
Fluorescence per RBC the sole sensitizing subclass in only five cases, and all
Hemolytic were blood donors. IgG3, thought by some investiga-
DAT+ Patients Anemia (n) Mean Range
tors to be the most reactive IgG subclass in immune
Idiopathic Yes (7) 99 32–255 RBC destruction, was present alone as commonly on
No (3) 35 32–41 the RBCs of patients without AIHA (6.3%) as on
Methyldopa-induced Yes (8) 57 28–127 patients with AIHA (6.6%), and was present on the
No (17) 40 30–53 RBCs of 2.6% of normal blood donors. Other investi-
Neonates* Yes (13) 63 30–205 gators have also detected IgG3 on RBCs of patients
No (25) 33 27–45 with no hemolytic anemia.102,106 These findings con-
*DAT+ babies born to mothers with antibodies of potential clinical significance
trast with the findings of Engelfriet and coworkers,107
who equated the presence of IgG3 with in vivo overt
hemolysis. It is of interest that multiple subclasses
were found more often in the patients with AIHA;
Qualitative Factors (e.g., Effect of IgG Class and later, Sokol and coworkers104 and Fabijanska-Mitek
Subclass). The Ig class of the antibody is very impor- and coworkers108 also reported this same association.
tant, as IgA does not activate complement or react Engelfriet and coworkers109 studied 746 patients
efficiently with most macrophages (see earlier). IgM is with positive DAT results due to IgG sensitization;
an efficient activator of complement, but does not unfortunately, no history of the presence or absence of
interact with macrophages as they have no IgM recep- hemolytic anemia was given. The percentage of
tor. If IgM antibodies do not destroy RBCs intravascu- patients with RBCs coated with Ig of each subclass,
larly, they can sensitize cells with complement, and alone or together with one or more other subclasses,
the cells may be removed extravascularly through C3 respectively, were: IgG1, 75/20%; IgG2, 0.7/11.7%;
macrophage receptor (CR1 and CR3) interactions. IgG3, 2.2/11%; and IgG4, 1/2.2%.

TABLE 4-10. IgG SUBCLASS OF AUTOANTIBODIES ON RBCs OF BLOOD


DONORS AND PATIENTS WITHOUT OR WITH AIHA6

Patients

Blood Donors % Without AIHA With AIHA


IgG Subclass (n = 89) (n = 32) (n = 46)

IgG1 alone 81.0 72.0 50.0


IgG2 alone 1.3 6.3 2.2
IgG3 alone 2.6 6.3 6.6
IgG4 alone 6.5 0.0 0.0
IgG1 with other subclasses 7.9 12.5 37.0
IgG2 with other subclasses 4.5 9.4 24.0
IgG3 with other subclasses 4.5 6.3 22.0
IgG4 with other subclasses 5.6 12.5 24.0
152 Immune Hemolytic Anemias

In 1982, Nance and Garratty110 made the surprising only IgM survive relatively normally. This finding
finding that IgG2 was detected alone on the RBCs of a correlates well with the fact that macrophages have no
patient with AIHA. As macrophages were originally receptor for IgM. Schreiber and Frank114 utilized a
reported not to have IgG2 receptors, this was of great strain of guinea pig genetically deficient in the fourth
interest. The patient was a 28-year-old woman of component of complement to show that RBCs sensi-
Hispanic origin with a diagnosis of AIHA. She tized with only IgM showed no accelerated clearance
responded favorably to steroid therapy. Her DAT whatsoever. Later experiments in humans with com-
result was positive due to sensitization with IgG (4+) plement deficiencies showed similar results to the
and complement (2+). Her serum contained warm guinea pig model.115 In contrast, Cutbush and
autoantibody (autoantibodies) reacting strongly (3+) Mollison116 and Burton and Mollison117 have
with all cells in the AHG phase with no apparent described examples of noncomplement-binding IgM
blood group specificity, and no alloantibodies were alloantibodies that led to a shortened RBC survival of
present. The eluate from her RBCs reacted strongly incompatible RBCs. It was suggested that this might
(4+) with all cells tested including –D– and Rhnull. IgG relate to agglutinates being trapped in small blood
subclassing of her RBCs showed that only IgG2 was vessels and eventually suffering metabolic damage. It
coating the cells. Monocyte monolayer assays using was pointed out that Schreiber and Frank114,115 used
the patient’s RBCs and allogeneic monocytes were relatively weakly sensitized RBCs in their experi-
strongly positive; there was 49% total reactivity ments. In retrospect, Mollison later admitted that the
(adherence and phagocytosis), the normal range being presence of IgG could not be excluded in his earlier
0 to 3%. In 1996, Roush and colleagues111 reported an experiments.7
unusual case of AIHA with reticulocytopenia, erythroid
dysplasia, and an IgG2 autoanti-U. Macrophages/
monocytes were originally reported not to possess DESTRUCTION OF RED CELLS SENSITIZED
IgG2 receptors, but now it is known that the WITH IgA ALONE
macrophages of individuals having a particular FcγRII
IgA serum alloantibodies and autoantibodies are
allotype can react efficiently with IgG2 (see p. 142).
uncommon, and for them to occur as the only
Like other investigators,112 the authors have not found
immunoglobulin class is rare. Nevertheless, several
IgG4 sensitization by itself to be associated with
examples of AIHA in which IgA autoantibodies alone
AIHA.
were shown to be present on the patient’s RBCs have
IgG3 appears to be more efficient in interacting with
been described.118-131 It is of interest to note that, in
mononuclear phagocytes than IgG1. Engelfriet and
one case studied by the authors (unpublished obser-
coworkers109 reported the following in vitro observa-
vations), a small number of IgG molecules were
tions: (1) IgG3– but not IgG1–weakly sensitized RBCs
demonstrated in addition to the IgA on the patient’s
could be cytoxically damaged in vitro when the DAT
RBCs, but only when they were tested by the sensi-
results on those RBCs was negative; (2) in vitro cyto-
tive antiglobulin consumption complement-fixation
toxic damage of IgG1-sensitized RBCs could be inhib-
assay (also see Chapter 9).132
ited more easily with IgG1 than IgG3-sensitized RBCs,
even when the IgG1-sensitized RBCs had more
RBC-bound IgG present. It may be important to note DESTRUCTION OF RED CELLS SENSITIZED
that all these experiments related to cytotoxicity and WITH IgG AND COMPLEMENT
not phagocytosis. They showed that, when RBCs were
weakly sensitized, then phagocytosis played a greater In the authors’ series of IHA, IgG and complement
role than cytotoxicity. The point at which phagocyto- were found together on the RBCs of 50% of the patients
sis became more important than cytotoxicity was at a (67% of the warm AIHA). It is difficult to prove that the
much lower degree of sensitization in the case of IgG3 IgG present on the RBC is responsible for the presence
than IgG1. Zupanska and colleagues113 showed that of the bound complement. Often, the IgG autoantibody
the minimum number of red cell–bound IgG mole- present in the serum or present in an eluate made from
cules needed for in vitro adherence to monocytes was the RBCs will not sensitize normal RBCs in vitro with
180 to 460 for IgG3 and 1180 to 4300 for IgG1. complement. Sometimes, complement sensitization can
be demonstrated by indirect antiglobulin test using an
eluate, even when fresh complement is not added to the
DESTRUCTION OF RED CELLS SENSITIZED eluate. It has been suggested that this can be explained
WITH IgM ALONE by the eluted IgG antibody carrying the complement
“piggyback.”133,134 It is possible that the presence of IgG
This situation rarely arises in vivo, as an IgM antibody is sometimes coincidental to the complement sensitiza-
usually activates complement when interacting with tion; complement may be activated by immune com-
the RBC, and the RBCs are then hemolyzed intravas- plexes remote from the RBCs (see Bystander Immune
cularly or removed extravascularly through C3b/iC3b Cytolysis in Chapter 9), or small amounts of IgM
macrophage interaction. Some interesting data have (undetectable by the antiglobulin test) may also be
been published suggesting that RBCs sensitized with present on the RBC surface and be responsible for the
Mechanisms of Immune Hemolysis 153

complement activation. All of the factors affecting 1. RBCs sensitized with C3 but not IgG were bound
destruction of RBCs sensitized only with IgG probably and formed rosettes with neutrophils. The rosettes
apply to cells sensitized with IgG and complement, frequently contained 10 or more RBCs per leuko-
together with the effects of complement sensitization. cyte; however, no RBCs were ingested.
There is experimental evidence in the literature sug- 2. Neutrophils required large amounts of IgG on the
gesting that extravascular RBC removal is enhanced RBCs to be stimulated for ingestion; 6 × 103 mole-
considerably when complement is present on the RBCs cules of IgG per RBC, without C3, induced no
in addition to IgG. Mollison and coworkers7 showed ingestion, and even with 6 × 104 molecules of IgG
that RBCs sensitized with 7 to 9 μg/mL of a comple- per RBC, only 70% of neutrophils ingested RBCs.
ment-binding IgG allo anti-Fya were cleared from the 3. C3 and Ig acted synergistically in opsonization.
circulation with a T50 of about 2 to 4 minutes (indicating With C3 bound to an IgG-sensitized RBC, ingestion
almost complete clearance of the cells at a single occurred with less than 10% of the amount of IgG
passage through the liver), whereas RBCs sensitized necessary for ingestion of an IgG-sensitized RBC
with a similar amount of noncomplement-binding IgG without C3.
antibody were cleared with a T50 on the order of 50
minutes, indicating negligible clearance by the liver In experiments with 125I-labeled C3, it was found that
and only partial clearance at each passage through the RBCs bearing 103 molecules of C3 together with 2 × 103
spleen. Schreiber and Frank115 injected normal guinea molecules of IgG were ingested by neutrophils more
pigs and guinea pigs deficient in complement with IgG- effectively than RBCs sensitized with 60 × 103 mole-
sensitized RBCs. The RBCs survived longer in animals cules of IgG alone. Similar results were obtained with
lacking either C4 or C3 and the terminal components, monolayers of human monocytes; that is, IgG was 10 to
suggesting that complement played a significant role in 30 times more effective in inducing phagocytosis when
determining the rate of destruction. RBCs were sensitized with C3. Monocytes, however,
Huber and colleagues44 were the first to show that showed a much greater sensitivity to opsonization with
human monocytes have a receptor for C3 and that it IgG alone. In many experiments, RBCs sensitized with
may function independently of IgG or cooperatively C3b and 102 molecules of IgG were ingested by 15% to
in the induction of phagocytosis. They showed that, if 45% of monocytes. The authors emphasized that the
the RBCs were sensitized with complement in addi- experiments just presented were carried out under con-
tion to IgG, then the inhibitory effect of free IgG, ditions in which the phagocytes were saturated with
described previously, was diminished considerably. In RBCs (overlay technique), and no shear forces existed
fact, in experiments in which RBCs sensitized with between RBC and phagocyte. The effect of C3 is
IgG only and IgG + C3 were added to monocytes, in magnified if the RBCs are kept in suspension (as they
the presence of IgG in the fluid phase, it could be would be in vivo). There is some evidence that C3d
shown that only 100 molecules of bound C3 per RBC may also play a synergistic role with IgG. Munn and
were sufficient to overcome the inhibitory effect of Chaplin140 showed that 1.5 to 5 times the amount of
IgG on the monocyte/sensitized RBC reaction. In adherence (rosetting) occurred in vitro when Rh
addition, C3 sensitization of the RBCs alone was (IgG)–sensitized RBCs had C3d present in addition to
found not to be sufficient to induce ingestion of RBCs IgG. Jaffe and coworkers141 also presented evidence to
by monocytes; when IgG antibodies were present, C3 support this theory; Rh (IgG)–sensitized RBCs were
appeared to initiate ingestion, suggesting a coopera- infused into normal volunteers and patients with a low
tive function in phagocytosis. Other workers135-138 serum C4 (0.1% of normal). Each of six patients with
have confirmed and extended these observations. low complement levels had a marked reduction in RBC
Mantovani and coworkers135 showed that, at 104 mol- clearance rates. They suggested that complement par-
ecules of IgG per RBC, binding of RBCs sensitized ticipates in Rh antibody–mediated clearance, and they
with IgG and complement was sixfold higher, but also demonstrated a synergistic role for C3d. Later
ingestion was only two to four times higher than with findings of a receptor for iC3b on macrophages45,48
IgG-only sensitized RBCs. These workers suggested suggest that this may have been responsible for the
that the receptors for IgG and C3 on the macrophage interactions credited to C3d in earlier reports.
membrane may not have the same function during It appears that the ability of particle-bound C3 to
phagocytosis, and that the C3 sites on the immune enhance phagocytosis can be explained solely by its
complexes are primarily involved in their attachment ability to bind the particle to the phagocyte. This is
to the macrophage, whereas the ingestion phase may also suggested by the observations that a variety of
depend mainly on the cytophilic receptor. The first agents that induce contact between particle and
contact between a phagocyte and the immune phagocyte, such as centrifugation, neuraminidase
complex is perhaps made through the C3 receptor, treatment of the RBCs, and the presence of dextran or
and the IgG receptor would be of primary importance protamine in the incubation medium, mimic the effect
only for interiorization. of C3. These experiments showed that, to be ingested
Ehlenberger and Nussenzweig139 studied the role of efficiently (and sometimes for ingestion to occur at
C3 and Fc receptors on human monocytes and neu- all), contact between particle and phagocyte must be
trophils. Some of their findings were: “boosted” by some agent. It was suggested that this
154 Immune Hemolytic Anemias

may be because most RBCs have an intrinsic negative cules of IgM per RBC (60 complement-fixing sites)
charge and can be expected to repel each other over were required to initiate immune clearance, whereas
short distances; overcoming two such barriers may although several thousand IgG molecules per RBC
represent a considerable problem in the recognition of were needed to form one C1 fixing site, as few as
a membrane-bound antibody molecule, or of other 1.4 IgG molecules per cell would effectively mediate
surface moieties capable of triggering phagocytosis by clearance. Complement sensitized RBCs are
the leukocytes. It was emphasized that, while C3 destroyed mainly in the liver.7,146,147 In contrast to
receptors serve to overcome this electrostatic repul- IgG, the complement receptor/complement sensi-
sion, the effect of C3 is clearly not mediated by a tized RBC interaction is not inhibited by plasma or
reduction of particle charge, as cells become even serum. This can be explained by the fact that the
more negatively charged after C3 is added. The receptor is specific for C3b and normal plasma or
enhancement of phagocytosis by particle-bound C3 is serum does not contain C3b which is, of course, an
specific; that is, it is due to its interaction with C3 activation product of C3. As the liver is a much larger
receptors of the plasma membrane of the phagocytes. organ with a larger blood flow, proportionally more
One important consequence of this concept is that C3 macrophage interactions will occur there, compared
may appear to trigger ingestion directly because with the spleen. Although all the experimental data
ingestion without C3 is negligible. Phagocytosis, in suggest that patients with IgG and complement on
these cases, may actually be triggered not by C3, but their RBCs should have more severe hemolysis than
by other moieties of the particle surface, which, in the those with equivalent amounts of IgG but not com-
absence of C3, are ineffective because of inefficient plement, this is not always so. The relationship
contact with the phagocyte. Such moieties, as, for between the presence of complement on RBCs, and
example, IgG in low doses, may function as “poten- the severity of autoimmune hemolysis is not clear.
tial” or “hidden” triggers of phagocytosis. It has been Some patients with IgG but no complement on their
suggested that the difference between IgG and C3 RBCs have more severe hemolysis than others with
with respect to their capacity to promote binding or cells strongly sensitized with complement and IgG.
ingestion may be more apparent than real and may Logue and Rosse148 found that, in general, those
depend on their respective surface distribution on the patients with both IgG and complement on the red
red cell membrane.142 Large amounts of IgG com- cell surface in large amounts have the greatest rates
bined with a high-density surface antigen are likely to of hemolysis. When the authors analyzed data from
be evenly distributed over the red cell membrane, their series, they found that there were many more
whereas C3 deposits in clusters around C42 sites.142 It patients with hemolytic anemia associated with
has been hypothesized that the distribution of the weak IgG sensitization (i.e., DAT anti-IgG titers
opsonic stimulus over the large area of the RBC mem- <1024) if complement was present on the RBCs than
brane determines the type of response by the phago- when it was absent.
cytic cell. It has been shown that the macrophage The authors have correlated the quantity of RBC–
membrane can ingest one opsonized particle while bound C3, serologic reactions, and hemolytic
failing to ingest a particle adjacent to it on the anemia.149 In vivo sensitized RBCs were obtained
macrophage membrane, but attached to the macro- from 25 patients who had a positive anti-C3 DAT
phage by a nonopsonic linkage [F(ab’)2 anti-F(ab’)2], result. None of 14 patients with fewer than 1000 mol-
that is to say, one lacking Fc.142 Thus, a phagocytic ecules of C3 per RBC had hemolytic anemia, whereas
stimulus does not appear to be propagated over the 8 of 11 patients with greater than 1000 molecules of C3
macrophage membrane, but may remain a local event. per RBC had overt hemolysis. The presence or
Specificity of the RBC–sensitizing antibody may also absence of hemolysis was not explained by variations
be important in determining the amount of in the amount of IgG on these patients’ RBCs. Thus,
macrophage interaction, as some RBC antigens may the amount of C3 per RBC was found to be an impor-
occur in clusters, whereas others may be more evenly tant determinant of hemolysis in vivo (see Chapter 5).
distributed over the membrane.143-145
It appears that macrophage C3 receptors can be acti-
vated by soluble stimuli (e.g., pharmacological and DESTRUCTION OF RED CELLS SENSITIZED
immunologic activators), and insoluble stimuli (e.g., WITH IgM AND COMPLEMENT (BUT NOT IgG)
plastic or glass). In resting macrophages, neither CR1
nor CR3 receptors initiate phagocytosis of C3-coated When RBCs are sensitized with IgM and complement
particles; however, some pharmacological activators is activated, frank hemolysis (i.e., intravascular hemol-
(e.g., the tumor promotor, phorbol myristate acetate) ysis) often occurs. Sometimes, the antibodies involved
can render both CR1 and CR3 competent to promote are not so readily hemolytic as, for instance, if group A
avid phagocytosis. blood is given to a group O hypogammaglobulinemic
Schreiber and Frank114,115 have shown that although patient with no anti-A detectable in vitro; the T50 of
IgG is less efficient in initiating complement fixation, it these RBCs may be 2 to 20 days with no gross sign of
appears to cause relatively more RBC damage than intravascular hemolysis.7 Some other alloantibodies,
IgM. In their studies in guinea pigs, at least 60 mole- such as anti-Lea, may also yield a similar pattern of
Mechanisms of Immune Hemolysis 155

destruction.7 Although there is no IgM receptor on ring C3 inactivator in plasma; the molecule is cleaved
macrophages, there are receptors for C3b and iC3b; into C3c, which is released into the plasma, and C3dg,
thus, if RBCs do not go all the way through the com- which remains attached to the RBC. The macrophage
plement cascade ending in lysis, they may be removed does not seem to interact efficiently, if at all, with C3dg
extravascularly if they have reached the C3 stage. in vivo. Thus, the RBC, although giving a strongly pos-
However, as discussed previously, this is an inefficient itive DAT due to bound complement (C3dg), may now
process in the absence of IgG. All of the factors dis- survive relatively normally. Schreiber and Frank114,115
cussed in the following section concerning the destruc- confirmed these data by using C4-deficient guinea pigs
tion of RBCs sensitized only with complement would and complement-deficient humans. Evidence has also
also play a role here. been presented in these publications that RBCs sensi-
tized only up to the C4 stage survive normally.
The presence of a C3d receptor on macrophages is
DESTRUCTION OF RED CELLS SENSITIZED ONLY controversial. Some workers140,152-154 have been able to
BY COMPLEMENT COMPONENTS detect a C3d receptor; others have not.136,137,146,155-157
Bianco157 makes the interesting observation that all the
Sometimes, RBCs can be sensitized with complement researchers who have detected a C3d receptor used
components and no immunoglobulins. The best purified complement components to prepare their sen-
example of this is associated with CAS. These patients sitized RBCs, whereas the researchers who could not
have a powerful IgM cold autoantibody present in their detect a receptor were using whole serum as a source of
plasma that reacts up to at least 30°C. When the patient complement. Bianco points out that the latter in vitro
is exposed to the cold and the peripheral circulation experiments would be closer to the in vivo situation;
falls to this temperature, the IgM antibody is able to certainly, all in vivo evidence so far would suggest that
react with the patient’s RBCs. The antibody activates the C3d receptor does not play an important role in the
complement, sometimes causing direct intravascular destruction of RBCs sensitized with C3d but not IgG;
lysis of some of the cells. The sensitized RBCs that however, as mentioned previously, recent reports have
escape lysis (in most cases, most of the RBCs escape suggested that C3d may play a synergistic role to IgG.
lysis) recirculate, and at 37°C, the cold antibody Some of the more recent findings on the structure of the
elutes from the RBCs into the plasma, leaving breakdown products of C3 suggest that some of the
complement firmly bound to the membrane. In earlier work may have related to the iC3b receptor on
1960, Lewis and coworkers150 showed that, when macrophages rather than C3d. C3b sensitization alone
complement-sensitized RBCs from a patient with CAS is a poor stimulus for phagocytosis.157-159 The adher-
were labeled with 51Cr and reinjected into the patient, ence of complement-sensitized RBCs to potentially
there was some immediate destruction of the RBCs; phagocytic cells requires few C3b molecules on the
however, most of the cells were sequestered by the liver membrane, but phagocytosis in vitro, even when
and later appeared back in the circulation and survived splenic macrophages are used, is distinctly uncommon.
normally. Mollison151 later obtained similar results after Brown and coworkers146 confirmed these in vitro
injecting volunteers with their own RBCs sensitized findings but found that, when such complement-sensi-
with complement in vitro by such methods as incuba- tized RBCs were injected into rabbits, evidence of
tion in low ionic strength solutions, etc.; these RBCs, phagocytosis occurred. Rosse and coworkers159 sug-
although having strongly positive DATs with anticom- gested that the number of complement molecules
plement sera, survived relatively normally. Brown and present on the membrane could affect the results. When
colleagues146 elaborated on these experiments by using more C3b molecules are present, engulfment is more
complement-deficient animals (C6-deficient rabbits). likely to occur; however, the number of C3b molecules
These workers again showed that if complement-sensi- needed for this reaction approaches the number that
tized cells (EAC43) do not react all the way through the leads to the direct lysis of the red cell, something on the
complement sequence to lysis, they become order of 100,000 C3b molecules per red cell. Both
sequestered in the liver and are later released and Brown146 and Rosse and coworkers159 believe that the
survive normally. It was shown that complement- distribution of C3b and IgG molecules on the red cell
sensitized RBCs attach to hepatic macrophages membrane may explain the differences in their reaction
(Kupffer cells) in the liver, presumably through the with phagocytic cells. The distribution of C3b is appar-
specific C3b macrophage receptor, where some are ently in spots of high concentration. Antibody, on the
immediately ingested. To activate the hepatic clearance other hand, is probably distributed randomly over the
mechanism, approximately 550 to 800 C3b molecules RBC surface.142 A more random distribution, as in the
per RBC are required. With time, unphagocytosed case of antibody, may serve as a greater stimulus to
RBCs return to the circulation at a slow exponential rate continued metapod activity in attempting engulfment,
(T50 of 25 to 100 minutes). It was suggested that the as compared with a more spotty distribution in the case
return of the EAC43 cells to the circulation from sites of of C3b.
attachment on fixed macrophages was the result of pro- An interesting observation is that, if C3b is deposited
gressive in vivo inactivation of fixed C3 (see Fig. 4-7). on the membrane adjacent to the I blood group anti-
The RBC-bound C3b is acted on by the naturally occur- genic site and then cleaved to C3d, the C3d remains on
156 Immune Hemolytic Anemias

the membrane in the form of an “immunologic scar.”160 gested that there might be two explanations for these
That is to say, eventually, C3d molecules occupy such a results: (1) there might be a subpopulation of mono-
large number of sites in the proximity of the I antigen cytes that selectively binds the sensitized RBCs, or
that newly formed C3b molecules are sterically pre- (2) the patient’s monocytes may have an increased
vented from binding, and the RBCs are protected from affinity for autologous immunoprotein or an increase
further damage mediated by C3b.138,161-163 Such RBCs in the number of Fc receptor sites that have developed
can be shown, in vitro, to be resistant to anti-I hemoly- a restricted specificity. These researchers further specu-
sis, compared with nonsensitized RBCs, but they are not lated that the antigen-antibody complex that results
resistant to lysis by other antibodies (e.g., anti-A). Jaffe when RBCs combine with antibody in immune hemo-
and coworkers141 performed red cell survival studies lysis effects a structured modification in the Fc portion
using 51Cr-labeled RBCs that were (1) maximally sensi- of the IgG molecule; this binding could result in a
tized with C3d using anti-I and then exposed to IgM unique conformation of that portion of the Fc that
anti-A in vitro, and (2) maximally sensitized with C3d binds to the monocyte. Gallagher and colleagues168
using anti-I and then re-exposed to anti-I and comple- showed that monocytes from four of six patients with
ment. The RBCs sensitized in the second stage with AIHA had increased in vitro phagocytic activity against
anti-I + complement survived normally, but the RBCs IgG-sensitized RBCs.
sensitized with IgM anti-A were destroyed rapidly. Using 51Cr-labeled, IgG-sensitized RBCs to study
These in vivo experiments confirm the in vitro finding RES function, Kelton169 studied nine patients receiving
that suggests that once RBCs become sensitized with methyldopa therapy; five of these had a positive DAT
C3d by anti-I autoantibody, they are then protected result (only one of these had an associated hemolytic
against further lysis by auto anti-I. It also may explain anemia), and four had a negative DAT result. The
why there are reports of transfused cells being patients without hemolytic anemia, regardless of their
destroyed more rapidly than the patient’s own “pro- DAT result, had significantly impaired RES clearance;
tected” RBCs in patients with CAS.164 in contrast, the one patient with hemolytic anemia did
In summary, complement sensitization of RBCs may not have impaired RES function (Fig. 4-18). Kelton169
result in (1) intravascular hemolysis; (2) sequestration suggested that methyldopa usually impairs macro-
in the reticuloendothelial system (e.g., liver) with phage function, and that is why so few patients with
ingestion by phagocytes; (3) sequestration with later methyldopa-induced positive DAT have associated
return to the circulation, where the red cell may have hemolysis. He suggested that, on rare occasions, an
normal survival; or (4) essentially normal survival. individual may have normal macrophage function and
thus will have an associated hemolytic anemia. Branch
and coworkers170 tested monocytes from two patients
VARIATION IN MACROPHAGE ACTIVITY with methyldopa-induced hemolytic anemia sequen-
tially over a 43- and 60-week period. No statistically
Variation in macrophage activity may well explain significant differences were noted between the patients’
some of the discrepant results observed in individual or normal donors’ monocyte activity (against IgG-sen-
patients (e.g., patients with severe hemolysis associated sitized RBCs).170
with weakly sensitized RBCs). MacKenzie165 found Frank and coworkers171 injected patients with and
that monocytes from patients with AIHA showed 40% without systemic lupus erythematosus (SLE) with
to 80% ingestion of IgG-sensitized RBCs, compared 51Cr-labeled RBCs strongly sensitized with anti-D.
with 10% to 15% when monocytes from normal donors Figure 4-19 shows the great differences in RBC sur-
were used. Patients with nonhemolytic anemias and vivals that were seen. The IgG-sensitized RBCs were
microangiopathic hemolytic anemia did not show rapidly destroyed in all patients without SLE, but in the
such differences. Kay and Douglas166 also studied SLE group, RBC survival varied from relatively normal
21 patients with immune hemolytic anemia; 4 were to rapid destruction. Frank and coworkers171 postu-
studied sequentially. Fifteen of 17 patients showed lated that the relatively good survival of these RBCs,
enhanced monocyte activity (i.e., rosetting and phago- strongly sensitized with IgG anti-D, was attributable to
cytosis) with autologous sensitized RBCs, as compared blockade of the RES (i.e., in the SLE patients, the Fc
with normal monocytes exposed to the same sensitized receptors of the macrophages were saturated with
RBCs. The most consistent elevation in monocyte activ- DNA/anti-DNA complexes), and thus could not inter-
ity occurred in RBCs from patients with RBCs sensi- act efficiently with the IgG-sensitized RBCs. It is not
tized with both IgG and complement. Of great interest hard to imagine that this phenomenon might occur
was the finding that there seemed to be no difference in with a wide range of immune complexes, in varying
activity between monocytes from patients or normal degrees, in many patients. It seems that this might be a
donors when tested against sheep RBCs sensitized with common explanation for different RBC survivals and
IgG rabbit antisheep (Forssman) antibody or IgM anti- clinical reactions when incompatible blood is trans-
body and complement. This contrasted strikingly with fused. It also illustrates how careful one should be
work by the same group showing increased IgG recep- about labeling an antibody of a certain specificity as
tor activity for patients with sarcoidosis, Crohn’s being nonpathogenic, when studies (e.g., 51Cr-labeled
disease, and tuberculosis.167 Kay and Douglas166 sug- RBC survival) in only one patient have been performed.
Mechanisms of Immune Hemolysis 157

100
>1500
90

80

700
70

60 600
Percent sensitized cells remaining in the circulation

T1/2 in minutes
50
500

40 400

300
30

200

100
20
50

Normals SLE

DAT + FIGURE 4-19. Clearance of IgG (anti-D)–sensitized RBCs in patients


NO HEMOLYSIS [ with active systemic lupus erythematosus (SLE) and in normal persons.
DAT –
(From Frank MM, Hamburger MI, Lawley TJ, Kimberly RP, Plotz PH:
Defective reticuloendothelial system Fc-receptor function in systemic
HEMOLYSIS DAT + lupus erythematosus. N Engl J Med 1979;300:518.)

10 20 30 60 4 normal subjects during the course of viral infections,


Time (minutes) with peak values exceeding 5, 8, 13, and 14 times the
normal mean. Elevations were also observed with
FIGURE 4-18. The clearance of IgG–sensitized, Cr-labeled, autologous monolayers prepared from blood of selected patients
RBCs in patients taking methyldopa. Four patients had positive direct
antibody test (DAT) results and no hemolysis (䊉—䊉), and four patients
with hematologic disorders. A remarkable value 168
had negative DAT results and no hemolysis (䊊—䊊). One patient had a times the normal mean was repeatedly obtained for a
positive DAT result and hemolytic anemia (䊉- - -䊉). The hatched area patient with myeloid metaplasia whose monolayer
represents the mean (± SD) clearance in 12 healthy controls. (From consisted almost exclusively of mature, granulocytic,
Kelton JG: Impaired reticuloendothelial function in patients treated with rosette-forming cells. The results emphasized the
methyldopa. N Engl J Med 1985;313:596.)
influence of monolayer donor variables on rosette for-
mation. There is considerable anecdotal evidence for
Munn and Chaplin140 studied factors affecting exacerbation of autoimmune hemolysis in relation to
donors of monocytes used in leukocyte monolayer intercurrent viral infections; the authors have cer-
activity testing involving 51Cr-labeled IgG Rh– tainly seen examples of this themselves. The findings
sensitized RBCs. Variation in rosette formation among of Munn and Chaplin140 would be consistent with
monolayers obtained from 10 healthy donors was such exacerbations, reflecting, at least in part, on acti-
defined, as was variation among 24 sequential mono- vation in vivo of the macrophage system.
layers obtained from one healthy donor and 8 sequen- Atkinson and Frank172 showed increased clearance
tial monolayers from a second healthy donor over a of IgG-sensitized RBCs in infected animals, hypothe-
4-month period. Rosette formation, expressed as sizing that increased macrophage activation in such
eluate cpm per plate, averaged 220 (range of 35 to 472) animals is responsible for the shortened in vivo RBC
for the 10 donors; 24 determinations on one donor survival. A substantial increase of Fc receptors (i.e.,
averaged 201 (range of 59 to 420), whereas 8 determi- five times greater than normal) on circulating
nations on the second donor averaged 244 (range of human monocytes has been shown to occur in indivi-
69 to 395). Striking increases in rosette formation were duals with solid malignant tumors.173 Arend and
observed employing monolayers made from 4 of Mannik174 demonstrated an approximate doubling of
158 Immune Hemolytic Anemias

IgG receptors and an increase in IgG association some workers have only demonstrated the in vitro
constants for rabbit alveolar macrophages following reactions with Rh-sensitized RBCs when the target
maximal stimulation with complete Freund’s adjuvant. RBCs were enzyme treated. Some investigators have
We and others2,175–184 have observed several cases of found that in vitro lymphocyte-induced ADCC,
AIHA following vaccination and immunization (see demonstrated with human blood group alloantibod-
Chapter 3), and it is tempting to speculate that activa- ies, was almost as efficient as that obtained with
tion of the macrophage system may set off increased monocytes. Kurlander and Rosse76 reported that, in
immune RBC destruction in patients whose RBCs may contrast to monocytes,89 purified IgG inhibited lym-
already have been sensitized with small quantities of phocyte-mediated ADCC, but human plasma did
protein and have been surviving relatively normally up not. They suggested that plasma contains a protein
to the time of vaccination. that augments lysis of sensitized RBCs by lympho-
Bianco and colleagues137 showed that both acti- cytes, despite high concentrations of unbound IgG. If
vated and nonactivated macrophages ingest IgG- this is true, then it suggests that lymphocyte-driven
coated RBCs (although activated cells ingested 1.5 to ADCC could play a significant role, sometimes, in
2 times as many), whereas nonactivated macrophages immune hemolysis.
avidly bind C3-coated RBCs but do not ingest them to
a significant degree. Activated macrophages, on the
other hand, bind and ingest C3-coated cells. NK CELLS
In 1981, Garratty185 suggested the possibility that NK
OTHER POSSIBLE MECHANISMS OF cells could recognize foreign determinants on RBCs
IMMUNE RED CELL DESTRUCTION and cause antibody-independent, cell-mediated, cel-
lular cytotoxicity. In one series, 6 of 22 patients with
Possible Role of Cells Other Than hemolytic transfusion reactions and no demonstrable
antibodies had chronic lymphatic leukemia. Garratty
Macrophages in Immune Red Cell and coworkers186 tried to demonstrate NK cell activity
Destruction using mononuclear cells from one patient with
LYMPHOCYTES chronic lymphatic leukemia against putative donor
RBCs. The experiment was unsuccessful, but Gilsanz
The role, if any, of lymphocytes in immune red cell and coworkers187 reported success in demonstrating
destruction is controversial. Lymphocytes are known NK cell activity against autologous RBCs in a patient
to have receptors for IgG (Fc) and complement (C3b, with DAT-negative AIHA. A 65-year-old patient with
C3d, and C4). They are known to be capable of a 5-year history of asymptomatic large granular lym-
destroying sensitized nucleated cells (e.g., other lym- phocytic leukemia developed hemolytic anemia. She
phocytes, tumor cells, and chicken RBCs), but their had a hemoglobin value of 97 g/L and an increased
role in the in vivo destruction of non-nucleated cells reticulocyte count (6%); the bilirubin value was
(e.g., RBCs) has been controversial. Before the target 1.4 mg/dL and the lactate dehydrogenase (LDH) level
cell is damaged, contact between the lymphocyte was 350 IU/L; serum haptoglobins were absent. The
and the target cell has to occur; complement may or peripheral blood smear showed marked spherocyto-
may not be involved. In vitro lymphocyte-driven sis; splenomegaly was found by ultrasonography.
lysis requires several hours to occur even under opti- DAT results were repeatedly negative. The large gran-
mal conditions. Cell-mediated cytotoxicity can be ular lymphocyte count was 6.0 X 109/L; the NK phe-
antibody-independent or -dependent (ADCC). When notype was CD2+, CD3-, CD16+, CD38+, CD57+. No
cell-mediated cytolysis by presensitized lympho- coexisting disease or hemolytic triggering factors
cytes occurs, the effector cells are thought to be cyto- were found. The patient did not improve with pred-
toxic T lymphocytes, and these do not require IgG nisone therapy (20 mg/day) and had to be transfused
antibodies as specific recognition factors for target twice for symptomatic anemia. The response to trans-
cell antigens. Cell-mediated cytotoxicity can also be fusions was good. To assess a possible role for NK
antibody-dependent (ADCC), in which case the cells in the hemolysis, a cytotoxicity assay was
target cells have to be sensitized with IgG antibody. designed. NK cells were obtained by density gradient
The effector cells for this reaction are thought to be and purified. Cytotoxicity was measured by a 51Cr
the so-called “killer” lymphocytes (K cells), which specific release assay using autologous RBCs and
are nonphagocytic and nonadherent lymphocytic ABO identical allogeneic RBCs as targets. The result-
cells. Some investigators believe K cells represent a ing hemolysis was expressed as a percentage of the
heterogenous family of effector cells (both B and T mean release, corrected for spontaneous release.
cells); others believe the effector cells are neither B Autologous RBCs, but not allogeneic RBCs, were
nor T cells. All are agreed that the cells possess an Fc lysed in vitro by the patient’s NK cells, either in the
receptor. It is now well documented that lympho- presence or absence of serum, indicating that a direct
cytes can cause in vitro lysis of RBCs sensitized with lytic mechanism was effected by the proliferating and
ABO and Rh antibodies.71-75 It should be noted that activated NK cells. The patient was started on treat-
Mechanisms of Immune Hemolysis 159

ment with cyclophosphamide. After 1 month, the logous). Recently, the same group presented evidence
anemia improved; 2 months later, NK blood cell to support this hypothesis.200 They were able to show
counts and spleen size normalized. Thereafter, the that “bystander” RBCs became sensitized in vitro with
patient’s NK cells failed to show any lytic activity complement by both the classical and alternative path-
against autologous RBCs in the cytotoxicity assay. It ways. For classical pathway activation, they used
was suggested that NK cell activation and cytotoxicity anti-A and group A RBCs; for alternative pathway
could have contributed to the hemolytic anemia in activation, they used human serum/Escherichia
this patient with large granular lymphocytic coli/human serum/rabbit RBCs and inulin/human red
leukemia.187 cells. They were able to demonstrate that bystander
RBCs acquired membrane-bound complement com-
ponents (e.g., group O cells became complement-
NEUTROPHILS sensitized when mixed with anti-A and group A RBCs).
The term “bystander lysis” has more recently been
There are very few data to suggest that neutrophils
applied to shortened survival of RBCs that become sen-
ever play a major role in RBC destruction. Zipursky
sitized with complement that is activated remote from
and Brown188 confirmed that normal neutrophils do
the putative RBCs. Petz201 has suggested that this form
not ingest IgG (Rh)–sensitized RBCs, but they were
of hemolysis may be more common than previously
able to show that neutrophils from patients with acute
recognized (see Chapter 9).
leukemia and leukocytosis readily ingest the sensi-
Reactive Hemolysis. The term “reactive hemolysis”
tized RBCs. Rosse and coworkers159 also confirmed
was coined by Thompson and Rowe202 in 1968 to
that normal granulocytes did not adhere to, or phago-
describe a form of bystander lysis that occurred in the
cytose, Rh (IgG)–sensitized RBCs, but they would
absence of RBC antibody and in the presence of ethyl-
adhere to and phagocytose RBCs sensitized with IgG
enediaminetetra-acetic acid (EDTA) (i.e., C1-inde-
anti-B. These findings suggest that an IgG receptor is
pendent lysis). Normal RBCs were lysed when
indeed present on granulocytes, but that the amount
incubated in sera to which antibody-coated bacteria or
of IgG present on the RBCs has to be considerable
other complement activators (e.g., zymosan and
before an interaction with the macrophage occurs.
agarose) had been added. These complement activa-
Sometimes, one can observe monocytes and/or gran-
tors are known to generate the C567 complex, without
ulocytes containing RBCs on peripheral blood smears
generating C42. This is similar to alternative pathway
from patients with AIHA, particularly in patients with
activation, but differs in that C3b is not detected on
PCH188–196 (see Chapter 3).
the cell. Lachmann and Thompson203 showed that,
when C567 is generated remote from the RBC, the
DESTRUCTION OF “INNOCENT BYSTANDER” complex can, if not inactivated (T50Cr = <1 minute),
RED CELLS (BYSTANDER LYSIS) attach to the RBCs. Such RBCs may hemolyze if they
take up C8 and C9. Götze and Mueller-Eberhard28,198
The complement cascade, once activated (e.g., by showed that RBCs from patients with PNH could be
immune complexes remote from the RBC), may possi- hemolyzed via a pathway that was independent of
bly cause complement sensitization and, indeed, lysis RBC antibody and the first four complement compo-
of non-Ig–sensitized “innocent bystander” RBCs (see nents. They showed that, once C42 was generated in
Chapter 9). The term “innocent bystander” was first the fluid phase or on a cell, C567 could attach to
coined by Dameshek,197 who applied it to RBCs that “bystanding” RBCs, which would lead to the adsorp-
were hemolyzed after adsorbing immune complexes. tion of C8 and C9 and subsequent lysis. C567 could
This was more readily demonstrable in vitro by using transfer from the site of activation on one cell to the
PNH RBCs.198 Later studies suggested that comple- site of binding on another cell. PNH RBCs, but not
ment sensitization may occur by a bystander mecha- normal RBCs, were lysed by this approach.27 A similar
nism with some regularity in delayed serologic and phenomenon had been described in 1965 by Yachnin
hemolytic transfusion reactions (DHTRs). Salama and and Ruthenberg,204 who termed it “indifferent activa-
Mueller-Eckhardt199 reported that all of 26 cases of tion.” Sirchia and coworkers205 showed that PNH
DHTR studied were associated with C3d sensitization RBCs were lysed, in vitro, in the presence of leuko-
of RBCs, even though many of the causative alloanti- cytes and leukocyte antibodies. They postulated that
bodies are usually thought to be incapable of comple- this reactive lysis of autologous RBCs was the cause of
ment activation (e.g., anti-Rh). The complement HTRs observed when patients with PNH were
sensitization was detectable on the RBCs for more than given compatible unwashed RBCs (i.e., RBCs with
3 months following the DHTR, and appeared to be sen- leukocytes present). In retrospect, as Sirchia and
sitizing autologous, as well as transfused, RBCs. They coworkers205 did not demonstrate that the lysis was
suggested alloantibody might activate the classical C3-independent, it might have been more correct to
pathway of complement and that fluid phase activation use the term “bystander lysis” rather than the more
of complement via the amplication of C3 convertase in specific term “reactive lysis.”
the alternative pathway may occur, leading to deposi- The possible role of bystander and/or reactive lysis
tion of C3b on “innocent bystander” RBCs (e.g., auto- in relation to DHTRs, post-transfusion crisis in sickle
160 Immune Hemolytic Anemias

cell disease, bone marrow transplantation, and post- enhanced (perhaps as a result of the binding of small
transfusion purpura is discussed in more detail in numbers of iC3b molecules to the RBCs). In contrast,
Chapters 9 and 14. the phagocytic and chemiluminescent responses of
unarmed monocytes to anti-D–sensitized RBCs were
abrogated in the presence of as little as 0.25% normal
ROLE OF ARMED MACROPHAGES IN IMMUNE serum, presumably as a result of the blocking of FcγRI
DESTRUCTION OF RED CELLS AND PLATELETS receptors, which have a high affinity for monomeric
IgG1 and IgG3. It was suggested that the first phase of
In 1960, Boyden and Sorkin206 showed that spleen cells the in vivo destruction of IgG-sensitized RBCs is the
from normal unimmunized rabbits that had been arming of macrophages by antibodies that are adsorbed
treated in vitro with antihuman albumin, made in onto macrophage FcγRI receptors. The adsorption of
rabbits or guinea pigs and washed, were capable of monomeric IgG is going on continuously, and RBC
specifically adsorbing 131I-labeled human albumin. antibodies are adsorbed along with the other mono-
They thought that this was due to a “cytophilic” anti- meric IgG molecules. Such armed macrophages would
body present in the rabbit and guinea pig sera. Some be capable of interacting with nonsensitized RBCs
recent observations may reflect the rediscovery of having the putative antigen (Fig. 4-20). The reactions
this observation. In 1985, Sunada and coworkers207 would be very efficient, as they are not inhibited by free
confirmed earlier reports that monocytes from patients IgG (and indeed may be enhanced by C3) in the plasma.
with AIHA phagocytosed anti-D–sensitized RBCs in The FcγRII and FcγIII receptors, which have a low
vitro more avidly than did monocytes from normal affinity for monomeric IgG, may react with antigen-
controls. When studying the factors affecting this inter- antibody complexes (i.e., IgG-sensitized RBCs), leading
action, they found that the monocytes from normal to the more commonly described opsonic reaction
controls could be made as active as the AIHA mono- between macrophages and sensitized RBCs.
cytes by incubating normal monocytes in antibody Another interesting aspect of this phenomenon is
eluted from the RBCs of patients with AIHA. Sunada the suggestion by Griffiths and coworkers209 that it
and coworkers207 used the term “armed” to describe may explain the destruction of RBCs when no anti-
the monocytes after incubation with autoantibody. body is readily demonstrable in the serum. It may be
In 1990, Hymes and coworkers208 reported that that all early antibody is adsorbed by the macro-
monocytes, incubated with an IgG fraction from phages, creating an efficient system for removal of
plasma of patients with autoimmune thrombocytope- nonsensitized RBCs by armed macrophages. At this
nia (ATP), bound to platelet glycoprotein (GP) IIb-IIIa stage of increased cell destruction, no antibodies may
(the major ATP autoantigen), immobilized on a
microtiter plate, 5.8 times more than normal mono-
cytes incubated with immobilized autoantigen opso-
nized with ATP IgG. Like Sunada and coworkers,207
Hymes and co-workers208 called the monocytes incu-
bated with ATP IgG armed monocytes. They showed
that the armed monocyte interaction required specific
F(ab’)2 antigen interaction. The greater efficiency of
the interaction of armed monocytes with autoantigen
compared with that of unarmed monocytes and
autoantibody-antigen (GPIIb-IIIa) complex was not
due to the quantity of antibody on the armed mono-
cytes, as six times as much IgG was present on the
GPIIb-IIIa antibody complex. The authors suggested
that the armed monocyte-platelet interaction, rather
than monocyte opsonized platelet interaction, may
be more pathophysiologically relevant in the destruc-
tion of platelets because opsonized platelets would
have to compete with plasma IgG for monocyte Fc
receptors.208
Griffiths and coworkers209 confirmed and extended FIGURE 4-20. A, Traditional view of extravascular RBC destruction. The
the findings reported by Sunada and coworkers207 Fc portion of the specific antibody attached to the RBCs reacts with
macrophage (M Fc (FcγRI, RII and/or RIII) receptors. B, “Armed”
Human monocytes were incubated with monoclonal macrophage: Monomeric IgG, which will include some specific RBC
anti-D and then incubated with D-positive RBCs. The antibodies, is adsorbed by macrophage FcRI receptors. The Fab
passively sensitized (armed) monocytes mediated portion of the “specific” RBC antibody can react with nonsensitized
inherent phagocytic and chemiluminescent responses. RBCs, leading to the same end product as the traditional pathway.
(From Garratty G: Novel mechanisms for immune destruction of
When the armed monocytes were incubated with D- circulating autologous cells. In Silberstein CE (ed): Autoimmune
positive RBCs in the presence of 50% normal serum, disorders of blood. Bethesda, Md: American Association of Blood
phagocytic and chemiluminescent responses were Banks, 1996, pp 79–114.)
Mechanisms of Immune Hemolysis 161

be detectable in the plasma. This may well explain the 3. Aschoff L: Ergebn inn. Med Kinderheilk 1924;26:1.
RBC survival collapse curve noted when small 4. van Furth R, Cohn ZA, Hirsch JG, et al: The mononuclear
phagocyte system: A new classification of macrophages,
volumes (<10 mL) of 51Cr-labeled RBCs are injected monocytes, and their precursor cells. Bull WHO 1972;46:845.
into previously nonimmunized individuals,7 but it is 5. Garratty G: The significance of complement in immunohema-
more difficult to extend this as an explanation for tology. CRC Crit Rev Clin Lab Sci 1984;20:25.
DAT-negative AIHA and hemolytic transfusion reac- 6. Garratty G: Factors affecting the pathogenicity of red cell auto-
tions with no detectable antibody. In these conditions, and alloantibodies. In Nance SJ (ed): Immune Destruction of
Red Blood Cells. Arlington, Va: American Association of Blood
one would think that antibody would eventually Banks, 1989:109.
appear in the plasma, but perhaps antibody could be 7. Mollison PL, Engelfriet CP, Contreras M: Blood Transfusion in
continually adsorbed by macrophages.210 Clinical Medicine, 10th ed. Boston, Mass: Blackwell Scientific,
1997.
8. Müller-Eberhard HJ: Complement. Ann Rev Biochem
IN VIVO AGGLUTINATION OF RED CELLS 1975;44:697–724.
9. Walport MJ: Complement. N Engl J Med 2001;344:1058–1067.
10. Borsos T, Rapp HR: Hemolysin titration based on fixation of
Before the role of the macrophage was fully appreci- the activated first component of complement: Evidence that
ated, aggregation or agglutination of RBCs in vivo one molecule of hemolysin suffices to sensitize an erythrocyte.
was believed to play a major role in immune RBC J Immunol 1965;95:559.
destruction. It was commonly believed that RBCs sen- 11. Humphrey JH, Dourmashkin RR: Electron microscope studies
sitized with nonagglutinating antibodies (e.g., IgG of immune cell lysis. In Wolstenholme GEW, Knight J (eds):
Complement. Boston: Little, Brown, 1965:175.
Rh) could be aggregated in the spleen where the 12. Humphrey JH, Dourmashkin RR: The lesions in cell mem-
protein concentration was high, and that these aggre- branes caused by complement. Adv Immunol 1969;11:75–115.
gates or agglutinates would be held up in the spleen 13. Mayer MM: Mechanism of cytolysis by complement. Proc Natl
and thus be more susceptible to destruction by Acad Sci USA 1972;69:2954–2958.
14. Pillemer L, Blum L, Lepow IH, et al: The properdin system and
macrophages. There is very little evidence to support immunity. I. Demonstration and isolation of a new serum
this theory as a major cause of immune RBC destruc- protein, properdin, and its role in immune phenomena.
tion, but it may play a role in some circumstances. Science 1954;120:279.
Romano and Mollison211 showed that RBCs coated 15. Nelson RA: An alternative mechanism for the properdin
with as little as 110 to 190 IgG anti-A molecules/RBC system. J Exp Med 1958;108:515.
16. Lepow IH: Louis Pillemer, properdin, and scientific contro-
would agglutinate if mixed with plasma and rocked versy. J Immunol 1980;125:471–475.
on a tile. By contrast, the minimum amount of IgG 17. Gewurz H, Shin HS, Mergenhagen SE: Interactions of the com-
anti-Rh required for agglutination under these condi- plement system with endotoxin lipopolysaccharides:
tions was about 19,000 IgG molecules/RBC. Consumption of each of the six terminal complement compo-
nents. J Exp Med 1968;128:1049–1057.
Complement-binding by RBCs coated with IgG 18. Schur PH, Becker EL: Pepsin digestion of rabbit and sheep
anti-A could be demonstrated only when the amount antibodies: The effect of complement fixation. J Exp Med
of antibody on the cells was at least 5134 IgG 1963;118:891.
molecules/RBC. Since, in ABO hemolytic disease, the 19. Sandberg AL, Oliveira B, Osler AG: Two complement interac-
amount of antibody on the cells is frequently less than tion sites in guinea pig immunoglobulins. J Immunol 1971;
106:282–285.
0.6 μg/mL (approximately 200 IgG molecules per red 20. Ellman L, Green I, Frank MM: Genetically controlled total
cell), it seems that RBC destruction in this syndrome is deficiency of the fourth component of complement in the
not caused by the activation of complement, but may guinea pig. Science 1970;170:74–75.
be due to the sequestration of agglutinated cells. 21. Frank MM, May JE, Gaither T, Ellman L: In vitro studies of
complement function in sera of C4 deficient guinea pigs. J Exp
Mollison and coworkers7 have also suggested that Med 1971;134:176–187.
some noncomplement-binding IgM alloantibodies, 22. Flexner S, Noguchi H: Snake venom in relation to hemolysis,
active at 37°C (e.g., examples of anti-c and anti-M), bacteriolysis, and toxicity. J Exp Med 1903;6:277.
may destroy RBCs in vivo because agglutinates may 23. Götze O, Müller-Eberhard HJ: The alternative pathway of
become trapped in small blood vessels and eventually complement activation. Adv Immunol 1976;24:1–35.
24. Schreiber RD, Pangburn MK, Lesavre PH, Müller-Eberhard
suffer metabolic damage. Such suggestions may also HJ: Initiation of the alternative pathway of complement:
be relevant to questions that are often asked about the Recognition of activators by bound C3b and assembly of the
potential of cold antibodies to cause clinical problems entire pathway from six isolated proteins. Proc Natl Acad Sci
in patients undergoing cardiac surgery using USA 1978;75:3948–3952.
25. Lachman PJ, Oldroyd RG, Milstein C, Wright BW: Three rat
hypothermia (see Chapter 9). monoclonal antibodies to human C3. Immunology 1980;41:503.
26. Lachman PJ, Pangburn MK, Oldroyd RG: Breakdown of C3
after complement activation. J Exp Med 1982;156:205–216.
27. May JE, Green I, Frank MM: The alternate complement
pathway in cell damage: Antibody-mediated cytolysis of ery-
R E F E R E N C E S throcytes and nucleated cells. J Immunol 1972;109:595–601.
28. Götze O, Müller-Eberhard HJ: Paroxysmal nocturnal hemoglo-
1. Fairley NH: The fate of extracorpuscular circulating haemo- binuria: Hemolysis initiated by the C3 activator system.
globin. Br Med J 1940;2:213. N Engl J Med 1972;286:180.
2. Dacie JV: The Haemolytic Anaemias, 2nd ed. Part II, The Auto- 29. Poskitt TR, Fortwengler HP, Lunskis BJ: Activation of the alter-
Immune Haemolytic Anemias. New York: Grune & Stratton, native complement pathway by autologous red cell stroma.
1962. J Exp Med 1973;138:715–722.
162 Immune Hemolytic Anemias

30. Turner MW: Mannose-binding lectin: The pluripotent mole- Immune-mediated Cell Destruction. Washington, DC:
cule of the innate immune system. Immunology Today 1996; American Association of Blood Banks, 1981:93.
17:532–540. 55. Holm G, Hammarstrom S: Haemolytic activity of human
31. Stahl PD, Ezckowitz RA: The mannose receptor is a pattern blood monocytes: Lysis of human erythrocytes treated with
recognition receptor involved in host defense. Curr Opin anti-A serum. Clin Exp Immunol 1973;13:29.
Immunol 1998;10:50–55. 56. Kurlander RJ, Rosse WF, Logue WL: Quantitative influence of
32. van Furth R: Origin and kinetics of monocytes and macro- antibody and complement coating of RBCs on monocyte-
phages. Semin Hematol 1970;7:125. mediated cell lysis. J Clin Invest 1978;61:1309–1319.
33. Huber H, Fudenberg HH: Receptor sites of human monocytes 57. Milgrom H, Shore SL: Lysis of antibody-coated human RBCs
for IgG. Int Arch Allergy 1968;34:18. by peripheral blood mononuclear cells. Cell Immunol
34. Huber H, Douglas SD, Nusbacher J, Kochwa S, Rosenfield RF: 1978;30:178–187.
IgG subclass specificity of human monocyte receptor sites. 58. Fleer A, Roos D, von dem Borne AEG Kr, Engelfriet CP:
Nature 1970;229:419. Cytotoxic activity of human monocytes towards sensitized
35. Abramson N, Gelfand EW, Jandl JH, Rosen FS: The interac- RBCs is not dependent on the generation of reactive oxygen
tion between human monocytes and RBCs. Specificity for IgG species. Blood 1979;54:407–411.
subclasses and IgG fragments. J Exp Med 1970;132:1207–1215. 59. Randazzo B, Hirschberg T, Hirschberg H: Cytotoxic effects of
36. van de Winkel JGJ, Capel PJA: Human IgG Fc receptor activated human monocytes and lymphocytes to anti-D-
heterogeneity: Molecular aspects and clinical implications. treated human erythrocytes in vitro. Scand J Immunol
Immunol Today 1993;14:215–221. 1979;9:351–358.
37. Ravetch JV, Bolland S: IgG Fc receptors. Ann Rev Immunol 60. Yust I, Goldsher N, Greenfeld R, Rabinovitz M: Hemolysis due
2001;19:275–290. to human antibody-dependent cell-mediated cytotoxicity.
38. Indik ZK, Park JG, Hunter S, Schreiber AD: The molecular Israel J Med Sci 1980;16:174–180.
dissection of Fcγ receptor mediated phagocytosis. Blood 61. Koller CA, LoBuglio AF: Monocyte-mediated antibody-
1995;86:4389–4399. dependent cell-mediated cytotoxicity: the role of the metabolic
39. Fanger MW, Shen L, Pugh J, Bernier GM: Subpopulations of burst. Blood 1981;58:293–299.
human peripheral granulocytes and monocytes express 62. Sagone AL Jr, Klassen DK, Decker MA, Clark L, Metz EN:
receptors for IgA. Proc Natl Acad Sci USA 1980;77:3640–3644. Characteristics of the metabolic response of human monocytes
40. Gauldie J, Richards C, Lamontagne L: Fc receptors for IgA to RBCs sensitized with anti-D alloantibodies. J Lab Clin Med
and other immunoglobulins on resident and activated alveo- 1981;98:382–395.
lar macrophages. Mol Immunol 1983;20:1029. 63. Todd RF III, Torchia RA, Peterson KE, Leeman EL: Binding and
41. Maliszewski CR, March CJ, Schoenborn MA, Gimpel S, lysis of antibody-coated human erythrocytes by activated
Shen L: Expression cloning of a human Fc receptor for IgA. J human monocytes. Clin Immunol Immunopathol 1984;30:
Exp Med 1990;172:1665–1672. 413–429.
42. Shen L: Receptors for IgA on phagocytic cells. Immunol Res 64. Yust I, Frisch B, Goldsher N: Antibody-dependent cell-
1992;11:273–282. mediated cytotoxicity (ADCC) of penicillin-treated human red
43. Morton HC, van Egmond K, van de Winkel JGJ: Structure and blood cells. Br J Haematol 1981;47:443–452.
function of human IgA Fc receptors (FcαR). Crit Rev Immunol 65. Yust I, Frisch B, Goldsher N: Antibody-dependent cell-
1996;16:423–440. mediated cytotoxicity and phagocytosis of autologous red
44. Huber H, Polley M, Linscott W, Fudenberg HH, blood cells in alphamethyldopa-induced haemolysis. Scand J
Muller-Eberhard H: Human monocytes: Distinct receptor Haematol 1986;36:211–216.
sites for the third component of complement and for 66. Samdal HH, Michaelsen TE, Heier HE, Nordhagen R:
immunoglobulin. G Science 1968;162:1281–1283. Antibody dependent cell mediated cytotoxicity against anti-D
45. Wright SD: Receptors for complement and the biology of sensitized human erythrocytes. APMIS 1988;96:250–256.
phagocytosis. In Gallin JI, Goldstein IM, Snyderman R (eds): 67. Fleer A, van der Hart M, von dem Borne AEG Kr,
Inflammation: Basic Principles and Clinical Correlates, 2nd Engelfriet CP: Monocyte-mediated lysis of human erythro-
ed. New York: Raven Press, 1992:477. cytes. In Eijsvoogel VP, Roos D, Zeimaker WP, eds:. Leucocyte
46. Sim RB, Walport MJ: C3 receptors. In Whaley K (ed): Membrane Determinants Regulating Immune Reactivity. New
Complement in Health and Disease. Boston: MTP Press Ltd, York: Academic Press, 1976:675.
1987:125. 68. Fleer A, van der Hart M, von dem Borne AEG Kr,
47. Cooper NR: Immune adherence by the fourth component of Engelfriet CP: Mechanism of antibody-dependent cytotoxicity
complement. Science 1969;165:396–398. by human blood monocytes towards IgG-sensitized erythro-
47a. Shibuya A, Sakamoto N, Shimizu Y, et al: Fc α/μ receptor cytes. Eur J Clin Invest 1976;6:333.
mediates endocytosis of IgM-coated microbes. Nat Immunol 69. Fleer A, Koopman MJ, von dem Borne AEG Kr, Engelfriet CP:
2000;1:441–446. Monocyte-induced increase in osmotic fragility of human
48. Moulds JM: Association of blood group antigens with RBCs sensitized with anti-D alloantibodies. Br J Haematol
immunologically important proteins. In Garratty G (ed): 1978;40:439–446.
Immunobiology of Transfusion Medicine. New York: Marcel 70. Fleer A, van Schaik MLJ, von dem Borne AEG Kr, Engelfriet
Dekker Inc., 1994:273. CP: Destruction of sensitized erythrocytes by human mono-
49. Lo Buglio AF, Cotran R, Jandl JH: Red cells coated with cytes in vitro. Effects of cytochalasin B, hydrocortisone and
immunoglobulin G: Binding and sphering by mononuclear colchicine. Scand J Immunol 1978;8:515–524.
cells in man. Science 1967;158:1582–1585. 71. Hinz CF Jr, Chickosky JF: Lymphocyte cytotoxicity for human
50. Brown DL, Nelson DA: Surface microfragmentation of RBCs erythrocytes. In Schwarz MR (ed): 6th Leucocyte Culture
as a mechanism for complement-mediated immune sphero- Conference, University of Washington, 1971. New York:
cytosis. Br J Haematol 1973;24:301. Academic Press, 1972:699.
51. Weed RI: The importance of erythrocyte deformability. Am J 72. Kurnick JT, Grey HM: Relationship between immunoglobulin-
Med 1970;49:147–150. bearing lymphocytes and cells reactive with sensitized human
52. Cooper RA: Loss of membrane components in the pathogen- erythrocytes. J Immunol 1975;115:305–307.
esis of antibody-induced spherocytosis. J Clin Invest 1972;51: 73. Shaw GM, Levy PC, LoBuglio AF: Human lymphocyte
16–21. antibody-dependent cell-mediated cytotoxicity (ADCC)
53. Mohandas N, de Boisfleury A: Antibody-induced spherocytic toward human red blood cells. Blood 1978;52:696–705.
anemia. I. Changes in red cell deformity. Blood Cells 1977;3:187. 74. Kurlander RL, Rosse WF, Ferreira E: Quantitative evaluation
54. Engelfriet CP, von dem Borne AEG Kr, Beckers D, et al: of antibody-dependent lymphocyte-mediated lysis of human
Immune destruction of RBCs. In Bell CA (ed): A Seminar on RBCs. Am J Hematol 1979;6:295–311.
Mechanisms of Immune Hemolysis 163

75. Lajos J, Benczur M, Nemák P: Characteristics of different group antibodies by the antiglobulin test. Vox Sang
Rhesus alloantibodies in lymphocyte dependent (K cell) cyto- 1964;9:385.
toxicity to human erythrocytes. Vox Sang 1979;36:240–243. 98. van der Meulen FW, de Bruin HG, Goosen PCM et al:
76. Kurlander RL, Rosse WF: Lymphocyte-mediated lysis of anti- Quantitative aspects of the destruction of RBCs sensitized with
body coated human RBCs in the presence of human serum. IgG1 autoantibodies: an application of flow cytofluorometry.
Blood 1979;53:1197–1202. Br J Haemat 1980;46:47.
77. Shaw GM, Aminoff D, Balcerzak SP, LoBuglio AF: Clustered 99. Garratty G, Nance SJ: Correlation between in vivo hemolysis
IgG on human red blood cell membranes may promote human and the amount of red cell-bound IgG measured by flow
lymphocyte antibody-dependent cell-mediated cytotoxicity. cytometry. Transfusion 1990;30:617-21.
J Immunol 1980;125:501–507. 100. Merry AH, Thomson EE, Rawlinson VI, Stratton F: A quantita-
78. Bakás T, Kimber I, Ringwald G, Moore M: K cell mediated tive antiglobulin test for IgG for use in blood transfusion
haemolysis: Influence of large numbers of unsensitized cells on serology. Clin Lab Haemat 1982;4:393.
the antibody-dependent lysis of anti-D-sensitized erythrocytes 101. Merry AH, Thomson EE, Rawlinson VI, Stratton F: Quantitation
by human lymphocytes. Br J Haematol 1984;57:447–455. of IgG on erythrocytes: correlation of number of IgG molecules
79. Sándor J, Lajos J, Braun J: Lymphocyte killer activity in indi- per cell with the strength of the direct and indirect antiglobulin
viduals sensitized against anti-D assessed by direct ADCC tests. Vox Sang 1984;47:73-81.
lysis of O, Rh-positive erythrocytes. Vox Sang 1986;51:310–313. 102. Dubarry M, Charron C, Habibi B, Bretagne Y, Lambin P:
80. Kumpel BM, Leader KA, Merry AH, et al: Heterogeneity in the Quantitation of immunoglobulin classes and subclasses of
ability of IgG1 monoclonal anti-D to promote lymphocyte- autoantibodies bound to RBCs in patients with and without
mediated red cell lysis. Eur J Immunol 1989;19:2283–2288. hemolysis. Transfusion 1993;33:466-71.
81. Holm G: Lysis of antibody-treated human erythrocytes by 103. Lynen R, Neuhaus R, Schwarz DWM, et al: Flow cytometric
human leukocytes and macrophages in tissue culture. Int Arch analyses of the subclasses of red cell IgG antibodies. Vox Sang
Allergy 1972;43:671. 1995;69:126-30.
82. Milgrom H, Shore SL: Lysis of antibody coated human RBCs 104. Sokol RJ, Hewitt S, Booker DJ, Bailey A: Red cell autoanti-
by peripheral blood mononuclear cells. Altered effector cell bodies, multiple immunoglobulin classes, and autoimmune
profile after treatment of target cells with enzymes. Cell hemolysis. Transfusion 1990;30:714-17.
Immunol 1978;30:178. 105. Sokol RJ, Hudson G: Quantiation of red cell-bound immuno-
83. Urbaniak SJ: Lymphoid cell dependent (K-cell) lysis of human protein. Transfusion 1998;38:782-95.
erythrocytes sensitized with rhesus alloantibodies. Br J 106. Sokol RJ, Hewitt S, Booker DJ, Bailey A: Erythrocyte autoanti-
Haematol 1976;33:409–413. bodies, subclasses of IgG and autoimmune haemolysis.
84. Northoff H, Kluge A, Resch K: Antibody dependent cellular Autoimmunity 1990;6:99-104.
cytotoxicity (ADCC) against human erythrocytes, mediated by 107. Engelfriet CP, von dem Borne AEG Kr, Beckers D, van Loghem
blood group alloantibodies: A model for the role of antigen JJ: Autoimmune haemolytic anaemia. Serological and
density in target cell lysis. Z Immun Forsch 1978;154:15. immunochemical characteristics of the autoantibodies:
85. Handwerger BS, Kay NW, Douglas SD: Lymphocyte-mediated Mechanisms of cell destruction. Ser Haematol 1974;8:
antibody-dependent cytolysis: Role in immune hemolysis. Vox 328–347.
Sang 1978;34:276–280. 108. Fabijanska-Mitek, Lopienska H, Zupanska B: Gel test applica-
86. von dem Borne AEG Kr, Engelfriet CP, Beckers D, van Loghem tion for IgG subclass detection in auto-immune haemolytic
JJ: Autoimmune haemolytic anaemia with incomplete warm anaemia. Vox Sang 1997;72:233–237.
autoantibodies. Clin Exp Immunol 1971;8:377–388. 109. Engelfriet CP, Overbeeke MAM, von dem Borne AEG Kr:
87. Fleer A, van der Meulen FW, Linthout E, von dem Borne AEG Autoimmune hemolytic anemia. Semin Hematol 1992;29:3.
Kr: Destruction of IgG-sensitized erythrocytes by human 110. Nance S, Bourdo S, Garratty G: IgG2 red cell sensitization asso-
blood monocytes: Modulation of inhibition by IgG. Br J ciated with autoimmune hemolytic anemia. Transfusion
Haematol 1978;39:425. (abstract) 1983;23:413.
88. Kelton JG, Singer J, Rodger C, et al: The concentration of IgG 111. Roush GR, Rosenthal NS, Gerson SL, et al: An unusual case of
in the serum is a major determinant of Fc-dependent reticu- autoimmune hemolytic anemia with reticulocytopenia, ery-
loendothelial function. Blood 1985;66:490–495. throid dysplasia, and an IgG2 autoanti-U. Transfusion
89. Kurlander RJ, Rosse WF: Monocyte-mediated destruction in 1996;36:575–580.
the presence of serum of RBCs coated with antibody. Blood 112. von dem Borne AEG Kr, Beckers D, Van der Meulen W,
1979;54:1131–1139. Engelfriet CP: IgG4 autoantibodies against erythrocytes,
90. Scornik JC, Salinas MC, Drewinko B: IgG dependent clearance without increased haemolysis: A case report. Br J Haematol
of red blood cells in IgG myeloma patients. J Immunol 1975; 1977;37:137.
115:901–903. 113. Zupanska B, Thomson EE, Merry AH: Fc receptors for IgG1 and
91. Constanoulakis M, Costea N, Schwartz RS, Dameshek W: IgG3 on human mononuclear cells—An evaluation with known
Quantitative studies of the effect of red blood cell sensitization levels of erythrocyte-bound IgG. Vox Sang 1986;50:97–103.
on in vitro hemolysis. J Clin Invest 1963;42:1790. 114. Schreiber AD, Frank MM: Role of antibody and complement in
92. Rosse WF: Correlation of in vivo and in vitro measurements of the immune clearance and destruction of erythrocytes. I. In
hemolysis in hemolytic anemia due to immune reactions. Prog vivo effects of IgG and IgM complement fixing sites. J Clin
Hematol 1973;8:51–75. Invest 1972;51:575–582.
93. Garratty G: The clinical significance (and insignificance) of 115. Schreiber AD, Frank MM: Role of antibody and complement in
red-cell-bound IgG and complement. In Wallace ME, Levitt JS the immune clearance and destruction of erythrocytes. II.
(eds): Current applications and interpretation of the direct Molecular nature of IgG and IgM complement fixing sites and
antiglobulin test. Arlington, Va: American Association of effects of their interaction with serum. J Clin Invest 1972;51:
Blood Banks, 1988:1. 583–589.
94. Garratty G: The significance of IgG on the red cell surface. 116. Cutbush M, Mollison PL: Relation between characteristics of
Trans Med Rev 1987;1:47. blood-group antibodies in vitro and associated patterns of red
95. Garratty G: Effect of cell-bound proteins on the in vivo sur- cell destruction in vivo. Br J Haematol 1958;4:115.
vival of circulating blood cells. Gerontology 1991;37:68–94. 117. Burton MS, Mollison PL: Effect of IgM and IgG iso-antibody
96. Horsewood P, Kelton JG: Macrophage-mediated cell destruc- on red cell clearance. Immunology 1968;14:861.
tion. In Garratty G (ed): Immunobiology of Transfusion 118. Engelfriet CP, von dem Borne AEG Kr, Giessen M, et al:
Medicine. New York: Marcel Dekker Inc., 1994:435–464. Autoimmune haemolytic anaemias. I. Serological studies with
97. Hughes-Jones NC, Polley MJ, Telford R, Gardner B, pure anti-immunoglobulin reagents. Clin Exp Immunol
Kleinschmidt G: Optimal conditions for detecting blood 1968;3:605–614.
164 Immune Hemolytic Anemias

119. Wager O, Haltia K, Rasauen JA, Vuopio P: Five cases of posi- 142. Griffin FM, Silverstein SC: Segmental response of the
tive antiglobulin test involving IgA warm type autoantibody. macrophage plasma membrane to a phagocytic stimulus. J Exp
Ann Clin Res 1971;3:76–85. Med 1974;139:323–336.
120. Stratton F, Rawlinson VI, Chapman SA, et al: Acquired 143. Nicholson GC, Masouredis SP, Singer SJ: Quantitative two
hemolytic anemia associated with IgA anti-e. Transfusion dimensional ultrastructural distribution of Rho (D) antigenic
1972;12:157–161. sites on human erythrocyte membranes. Proc Natl Acad Sci
121. Worlledge SM, Blajchman MA: The autoimmune haemolytic 1971;68:1416–1420.
anaemias. Br J Haematol 1972;23(S):61. 144. Masouredis SP, Sudora EJ, Mahan L, Victoria EJ: Antigen site
122. Sturgeon P, Smith LE, Chun HMT, et al: Autoimmune densities and ultrastructural distribution patterns of red cell
hemolytic anemia associated exclusively with IgA of Rh Rh antigens. Transfusion 1976;16:94–106.
specificity. Transfusion 1979;19:324–328. 145. Victoria EJ, Muchmore EA, Sudora EJ, Masouredis SP: The role
123. Suzuki S, AmanoT, Mitsunaga M, et al: Autoimmune of antigen mobility in anti-Rh (D) induced agglutination. J Clin
hemolytic anemia associated with IgA autoantibody. Clin Invest 1975;56:292–301.
Immunol Immunopathol 1981;21:247–256. 146. Brown DL, Lachmann PJ, Dacie JV: The in vivo behavior of
124. Wolf CFW, Wolf DJ, Peterson P, et al: Autoimmune hemolytic complement-coated RBCs: Studies in C6-deficient, C3-depleted
anemia with predominance of IgA autoantibody. Transfusion and normal rabbits. Clin Exp Immunol 1970; 7:401–421.
1982;22:238–240. 147. Brown DL: The behavior of phagocytic cell receptors in rela-
125. Clark DA, Dessypris EN, Jenkins DE Jr, Krantz SB: Acquired tion to allergic red cell destruction. Ser Haematol 1974;7:3.
immune hemolytic anemia associated with IgA erythrocyte 148. Logue G, Rosse W: Immunologic mechanisms in autoimmune
coating: Investigation of hemolytic mechanisms. Blood hemolytic disease. Semin Hematol 1976;113:277.
1984;64:1000–1005. 149. Fischer J, Petz LD, Garratty G, Cooper N: Correlations
126. Kowal-Vern A, Jacobson P, Okuno T, Blank J: Negative direct between quantitative assay of red cell bound C3, serologic
antiglobulin test in autoimmune hemolytic anemia. Am J reactions, and hemolytic anemias. Blood 1974;44:359–373.
Pediatr Hematol Oncol 1986;8:349–351. 150. Lewis SM, Dacie JV, Szur L: Mechanisms of haemolysis in the
127. Reusser P, Osterwalder B, Burri H, Speck B: Autoimmune cold-haemagglutinin syndrome. Br J Haematol 1960;6:164.
hemolytic anemia associated with IgA—Diagnostic and thera- 151. Mollison PL: The role of complement in haemolytic processes
peutic aspects in a case with long-term follow-up. Acta in vivo. In Wolstenholme GEW, Knight J (eds): Complement.
Haematol 1987;77:53–56. London: Churchill, 1965:323.
128. Girelli G, Perrone MP, Adorno G, et al: A second example of 152. Wellek B, Hahn HH, Opferkuch W: Evidence for macrophage
hemolysis due to IgA autoantibody with anti-e specificity. C3d-receptor active in phagocytosis. J Immunol 1975;114:
Haematologica 1990;75:182–183. 1643–1645.
129. Göttsche B, Salama A, Mueller-Eckhardt C: Autoimmune 153. Reynolds HY, Atkinson JP, Newball HH, Frank MM: Receptors
hemolytic anemia associated with an IgA autoanti-Gerbich. for immunoglobulin and complement on human alveolar
Vox Sang 1990;58:211–214. macrophages. J Immunol 1975;114:1813–1819.
130. Sokol RJ, Booker DJ, Stamps R, et al: Autoimmune hemolytic 154. Ehlenberger AG, Nussenzweig V: Synergy between receptors
anemia due to IgA class autoantibodies. Immunohematology for Fc and C3 in the induction of phagocytosis by human
1996;12:14–19. monocytes and neutrophils. Fed Proc Fed Am Soc Exp Biol
131. Sokol RJ, Booker DJ, Stamps R, Booth JR, Hook V: IgA red cell 1975;34:854.
autoantibodies and autoimmune hemolysis. Transfusion 155. Atkinson JP, Frank MM: Studies on the in vivo effects of anti-
1997;37:175–181. body interaction of IgM antibody and complement in the
132. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias, immune clearance and destruction of erythrocytes in man.
1st ed. New York: Churchill Livingstone, 1980. J Clin Invest 1974;54:339–348.
133. Gilliland BC, Leddy JP, Vaughan JH: The detection of 156. Müller-Eberhard HJ: Complement and phagocytosis. In
cell-bound antibody on complement-coated human RBCs. Bellanti JA, Delbert HD (eds): The Phagocytic Cell in Host
J Clin Invest 1970;49:898–906. Resistance. New York: Raven Press, 1975:87.
134. Leddy JP, Bakemeier RF, Vaughan JH: Fixation of complement 157. Bianco C: Plasma membrane receptors for complement. In
components to autoantibody eluted from human RBC. J Clin Good RA, Day SB (eds): Comprehensive Immunology. New
Invest 1965;44:1066. York: Plenum Press, 1975:69.
135. Mantovani B, Rabinovitch M, Nussenzweig V: Phagocytosis of 158. Griffin FM Jr, Griffin JA, Leider JE, Silverstein SC: Studies on
immune complexes by macrophages: Different roles of the the mechanism of phagocytosis. I. Requirements for circum-
macrophage receptor sites for complement (C3) and for ferential attachment of particle-bound ligands to specific
immunoglobulin (IgG). J Exp Med 1972;135:780–792. receptors on the macrophage plasma membrane. J Exp Med
136. Griffin FM, Jr, Bianco C, Silverstein SC: Characterization of the 1975;142:1263–1282.
macrophage receptor for complement and demonstration of its 159. Rosse WF, De Boisfleury A, Bessis M: The interaction of phago-
functional independence from the receptor for the Fc portion cytic cells and RBCs modified by immune reactions.
of immunoglobulin G. J Exp Med 1975;141:1269–1277. Comparison of antibody and complement coated RBCs. Blood
137. Bianco C, Griffin FM Jr, Silverstein SC: Studies of the Cells 1975;1:345.
macrophage complement receptor. J Exp Med 1975;141: 160. Atkinson JP, Frank MM: Role of complement in the pathophy-
1278–1290. siology of hematologic diseases. Prog Hematol 1977;10:
138. Scornik JC, Drewinko B: Receptors for IgG and complement in 211–245.
human spleen lymphoid cells: Preferential binding of parti- 161. Boyer JT: Complement and cold agglutinins. II. Interactions of
culate immune complexes through complement receptors. the components of complement and antibody within the
J Immunol 1975;115:1223–1226. haemolytic complex. Clin Exp Immunol 1967;2:241–252.
139. Ehlenberger AG, Nussenzweig V: Immunologically- 162. Engelfriet CP, von dem Borne AEG Kr, Beckers D, Reynierse E,
mediated phagocytosis: Role of C3 and Fc receptors. In van Loghem JJ: Autoimmune haemolytic anaemias. V. Studies
Litwin SD, Christian CL, Siskind GW (eds): Clinical on the resistance against complement haemolysis of the RBCs
Evaluation of Immune Function in Man. New York: Grune & of patients with chronic cold agglutinin disease. Clin Exp
Stratton, 1976:47. Immunol 1972;11:255–264.
140. Munn LR, Chaplin H JY, Jr: Rosette formation by sensitized 163. Evans RS, Turner E, Bingham M: Chronic hemolytic anemia
human RBCs—Effects of source of peripheral leukocyte mono- due to cold agglutinins: The mechanism of resistance of RBCs
layers. Vox Sang 1977;33:129–142. to C’ hemolysis by cold agglutinins. J Clin Invest 1967; 46:
141. Jaffe CJ, Atkinson JP, Frank MM: The role of complement in the 1461–1474.
clearance of cold agglutinin-sensitized erythrocytes in man. 164. Evans RS, Turner E, Bingham M: Studies with radio iodinated
J Clin Invest 1976;58:942–949. cold agglutinins of ten patients. Am J Med 1965;38:378.
Mechanisms of Immune Hemolysis 165

165. MacKenzie MR: Monocytic sensitization in autoimmune 189. Gelfand EW, Abramson N, Segel GB, Nathan DG: Buffy-coat
hemolytic anemia. Clin Res (abstract) 1975;23:132. observations and red-cell antibodies in acquired hemolytic
166. Kay NE, Douglas SD: Monocyte-erythrocyte interaction in anemia. N Engl J Med 1971;284:1250–1252.
vitro in immune hemolytic anemias. Blood 1977;59:889–897. 190. Spiva DA, George JN, Sears DA: Acute autoimmune hemolytic
167. Douglas SD, Schmidt ME, Siltzbach HE: Monocyte receptor anemia due to a low molecular weight IgM cold hemolytic
activity in normal individuals and patients with sarcoidosis. associated with episodic lymphoid granulomatous vasculitis.
Immunol Commun 1972;1:25–38. Am J Med 1974;56:417–428.
168. Gallagher MT, Branch DR, Mison A, Petz LD: Evaluation of 191. Djaldetti M, Elion D, Bessler H, Fishman P: Paroxysmal cold
reticuloendothelial function in autoimmune hemolytic anemia hemoglobinuria. Am J Clin Pathol 1975;63:804–810.
using an in vitro assay of monocyte-macrophage interaction 192. Wolach B, Heddle N, Barr RD, et al: Transient Donath-
with erythrocytes. Exp Hematol 1983;11:82–89. Landsteiner haemolytic anaemia. Br J Haematol 1981;48:425–434.
169. Kelton JG: Impaired reticuloendothelial function in patients 193. Lau P, Sererat S, Moore V, McLeish K, Alousi M: Paroxysmal
treated with methyldopa. N Engl J Med 1985;313:596. cold hemoglobinuria in a patient with Klebsiella pneumonia.
170. Branch DR, Gallagher MT, Shulman IA, et al: Reticulo- Vox Sang 1983;44:167–172.
endothelial cell function in α-methyldopa-induced hemolytic 194. Hernandez JA, Steane SM: Erythrophagocytosis by segmented
anemia. Vox Sang 1983;45:278–287. neutrophils in paroxysmal cold hemoglobinuria. Am J Clin
171. Frank MM, Hamburger MI, Lawley TJ, Kimberly RP, Plotz PH: Pathol 1984;81:787–789.
Defective reticuloendothelial system Fc-receptor function in 195. Uzokwe CO, Gwynn AM, Gorst DW, Adamson AR: Infectious
systemic lupus erythematosus. N Engl J Med 1979;300:518. mononucleosis complicated by haemolytic anaemia due to the
172. Atkinson JP, Frank MM: The effect of bacillus calmette-guerin- Donath Landsteiner antibody and by severe neutropenia. Clin
induced macrophage inactivation on the in vivo clearance of Lab Haematol 1993;15:137.
sensitized erythrocytes. J Clin Invest 1974;53:1742–1749. 196. Friedman HD, Dracker RA: Intravascular erythrophagocyto-
173. Rhodes J: Altered expression of human monocyte Fc receptors sis. JAMA 1991;265:1082.
in malignant disease. Nature 1977;265:253–255. 197. Dameshek W: Autoimmunity: Theoretical aspects. Ann NY
174. Arend WP, Mannik M: Quantitative studies on IgG receptors Acad Sci 1965;124:6–28.
on monocytes. In van Furth R (ed): Mononuclear Phagocytes 198. Götze O, Muller-Eberhard HJ: Lysis of erythrocytes by comple-
in Immunity, Infection, and Pathology. Oxford: Blackwell ment in the absence of antibody. J Exp Med 1970;132:898–915.
Scientific, 1975:303. 199. Salama A, Mueller-Eckhardt C: Delayed hemolytic transfusion
175. Kölbl H: Klinik und therapie der akuten erworbenen reactions. Transfusion 1984;24:188–193.
hämolytischen anämien im kindesalter. Ostdeutsch Z 200. Salama A, Mueller-Eckhardt C: Binding of fluid phase C3b to
Kinderheilk 1955;11:27. nonsensitized bystander human RBCs: A model for in vivo
176. Dameshek W, Schwartz R: Hemolytic Mechanisms. Ann NY effects of complement activation on blood cells. Transfusion
Acad Sci 1959;77:589. 1985;25:528–534.
177. Gedikoglu AG, Cantez T: Haemolytic anaemia relapses after 201. Petz LD: The expanding boundaries of transfusion medicine. In
immunisation and pertussis. Lancet 1967;2:894–895. Nance SJ (ed): Clinical and Basic Science Aspects of Immuno-
178. Pirofsky B: Infectious disease and autoimmune hemolytic hematology. Arlington, Va: American Association of Blood
anemia. In Pirofsky B (ed): Autoimmunization and the Banks, 1991:73.
Autoimmune Hemolytic Anemias. Baltimore: Williams & 202. Thompson RA, Rowe DS: Reactive haemolysis—A distinctive
Wilkins, 1969:153. form of red cell lysis. Immunology 1968;14:745.
179. Bossi E, Wagner HP: Autoimmune hemolytic anemia and 203. Lachmann PJ, Thompson RA: Reactive lysis: The complement-
cytomegalovirus infection in a six month old child, treated mediated lysis of unsensitized cells. II. The characteristics of
with azathioprine. Helv Paediat Acta 1972;27:155-62. activated reactor as C56 and the participation of C8 and C9.
180. Rosse WF: Correlation of in vivo and in vitro measurements of J Exp Med 1970;131:643–657.
hemolysis in hemolytic anemia due to immune reactions. Prog 204. Yachnin S, Ruthenberg JM: The initiation and enhancement of
Hematol 1973;8:51–75. human red cell lysis by activators of the first component of
181. Bunch CF, Schwartz CM, Bird GWG: Paroxysmal cold haemo- complement and by first component esterase; studies using
globinuria following measles immunization. Arch Dis Child normal RBCs and RBCs from patients with paroxysmal noc-
1975;47:299. turnal hemoglobinuria. J Clin Invest 1965;44:518.
182. Zupanska B, Lawkowicz W, Górska B, et al: Autoimmune 205. Sirchia G, Ferrone S, Mercuriali F: Leukocyte antigen-antibody
hemolytic anemia in children. Br J Haematol 1976;34: reaction and lysis of paroxysmal nocturnal hemoglobinuria
511–520. erythrocytes. Blood 1970;36:334–336.
183. Haneberg B, Matre R, Winsnes R, et al: Acute hemolytic 206. Boyden SV, Sorkin E: The adsorption of antigen by spleen cells
anemia related to diptheria-pertussis-tetanus vaccination. previously treated with antiserum in vitro. Immunology
Acta Paediatr Scand 1978;67:345–350. 1960;3:272.
184. Johnson ST, McFarland JG, Casper JT, Oshima S, Gottschall JL: 207. Sunada M, Suzuki S, Ota Z: Reticuloendothelial cell function
An unusual case of autoimmune hemolytic anemia following in autoimmune hemolytic anemia (AIHA): Studies on the
DPT vaccination (abstract). Transfusion 1989;29:50S. mechanism of peripheral monocyte activation. Acta Med
185. Garratty G: Basic mechanisms of in vivo cell destruction. In Okayama 1985;5:375–384.
Bell CA (ed): A Seminar on Immune-mediated Cell Destruc- 208. Hymes KB, Schuck MP, Karpatkin S: Regulation of autoim-
tion. Washington, DC: American Association of Blood Banks, mune anti-platelet antibody-mediated adhesion of monocytes
1981:1. to platelet GPIIb/BPIIIa: effect of armed monocytes and the
186. Garratty G, Vengelen-Tyler V, Postoway N, et al: Hemolytic Mac-1 receptor. Blood 1990;75:1813–1819.
transfusion reactions (HTR) associated with antibodies not 209. Griffiths HL, Kumpel BM, Elson CJ, Hadley AG: The func-
detectable by routine procedures (abstract). Transfusion tional activity of human monocytes passively sensitized with
1982;22:429. monoclonal anti-D suggests a novel role for FcγRI in the
187. Gilsanz F, de la Serna FJ, Moltó L, Alvarez-Mon M: Hemolytic immune destruction of blood cells. Immunology 1994;83:370.
anemia in chronic large granular lymphocytic leukemia of 210. Garratty G: Novel mechanisms for immune destruction of cir-
natural killer cells: Cytotoxicity of natural killer cells against culating autologous cells. In Silberstein CE (ed): Autoimmune
autologous RBCs is associated with hemolysis. Transfusion disorders of blood. Bethesda, Md: American Association of
1996;36:463–466. Blood Banks, 1996:79–114.
188. Zipursky A, Brown EJ: The ingestion of IgG-sensitized ery- 211. Romano EL, Mollison PL: Red cell destruction in vivo by low
throcytes by abnormal neutrophils. Blood 1974;43:737–742. concentrations of IgG anti-A. Br J Haematol 1975;29:121–127.
C H A P T E R 5

Differential Diagnosis
of Immune Hemolytic
Anemias

As indicated in Chapter 2, aspects of the autoantibodies, including their immu-


the laboratory evaluation of a noglobulin class and subclass and the immunochem-
patient who has an acquired istry of cold autoantibodies.
hemolytic anemia should
include a direct antiglobulin
test (DAT), which, if positive,
DISTINCTIVE CLINICAL AND ROUTINE
allows a presumptive diagnosis LABORATORY FEATURES
of an immune hemolytic
anemia (IHA). The physician Some of the clinical and routine laboratory findings in
must then differentiate among patients with IHAs are sufficiently distinctive as to
the various causes of IHA, which are listed in Table 3-1. strongly suggest the type of IHA that is present. The
We begin the discussion of the differential diagnosis following observations merit particular emphasis
by emphasizing distinctive clinical and laboratory because they are most helpful.
findings. The approach to the laboratory diagnosis of
each kind of autoimmune hemolytic anemia (AIHA) Association with Exposure to Cold
includes a narrative overview that omits technical
details but explains the significance of the results of A history of acrocyanosis and/or hemoglobinuria on
the tests described in Chapter 6. We discuss results of exposure to cold in an elderly patient with an
screening tests, more definitive tests, and characteris- acquired hemolytic anemia strongly suggests a diag-
tic findings in patients with IHAs of various types nosis of CAS. However, although reports in the early
(i.e., warm antibody AIHA (WAIHA), cold agglutinin medical literature on CAS stressed these disease
syndrome (CAS), paroxysmal cold hemoglobinuria manifestations, we find that they are absent in a
(PCH), and combined cold and warm AIHA). This majority of patients. Their absence in most patients
includes a review of some atypical serologic findings with CAS who have been diagnosed in recent years
such as AIHA caused by IgM warm autoantibodies or perhaps is attributable to the fact that CAS was the
IgG cold autoantibodies. (AIHA associated with a last type of AIHA to be well characterized, and, ini-
negative DAT is discussed in Chapter 9.) Although the tially, only those patients with the most severe disease
differential diagnosis of IHAs is based on clinical and were so diagnosed. A review of earlier serologic
serologic findings, we also review some basic science studies supports this impression. In Dacie’s1 series of

167
168 Immune Hemolytic Anemias

26 patients published in 1962, all but 2 had cold agglu- history of drug ingestion, which, in some instances,
tinin titers of 4000 or greater, whereas in our series, may have occurred a week or more before the onset of
43% of patients had a cold agglutinin titer less than hemolysis, or be a single dose given for surgery (e.g.,
1000 (see later). cefotetan). Knowledge of the drugs that have been
Although one might assume that PCH is commonly implicated as a cause of drug-induced IHA is essential
precipitated by exposure to cold, this is only occasion- (see Chapter 8).
ally true. Indeed Wolach and coworkers2 pointed out Many drug-induced IHAs can be distinguished
that the most common form of PCH is the transient from AIHA by laboratory findings, that is, the demon-
type, secondary to infection (e.g., in childhood), and stration of a drug-dependent red blood cell (RBC)
this is rarely paroxysmal, is only occasionally clearly antibody. However, the administration of some drugs
precipitated by cold, and is not invariably expressed causes hemolytic anemia that is serologically indistin-
as hemoglobinuria (although the latter finding is very guishable from cases of idiopathic WAIHA. We refer
common).3 to the latter cases as drug-induced AIHA, and cases
wherein a drug-dependent antibody can be identified
Autoagglutination we refer to as drug-induced IHA. The most common
drug at present to cause AIHA is fludarabine (see
Autoagglutination is a finding that may be noted by Chapter 8).
technologists in all sections of the laboratory, not just
those in the blood transfusion or immunohematology Alloantibody-Induced Immune
laboratories. Indeed, cold autoagglutinins that react
strongly at room temperature cause such striking
Hemolytic Anemia
findings that they are difficult to ignore. Auto- Alloantibody-induced hemolytic anemias include
aggutination visible to the naked eye occurring at hemolytic disease of the newborn and hemolytic
room temperature is characteristic of CAS, but may transfusion reactions. The clinical setting usually
also be noted in about one third of patients with strongly suggests these diagnoses. Although autoanti-
WAIHA.4 Although the autoagglutination caused by body-induced hemolytic disease of the newborn can
such cold agglutinins is often 2+ to 4+, it almost occur as a result of transplacental passage of a
always completely disperses after a few minutes of mother’s IgG warm autoantibody, this is very unlikely
incubation at 37°C (Fig. 5-1), whereas that caused by unless the mother has obvious and quite severe
warm autoantibodies is usually much weaker and WAIHA (see Chapter 9).
will not disperse at 37°C. If the blood sample has been Also, when hemolysis occurs in the immediate
obtained from a patient known to have hemolytic aftermath of an RBC transfusion, the diagnosis of an
anemia, such simple observations offer an important acute hemolytic transfusion reaction is quite evident.
clue to the correct diagnosis. However, distinguishing a delayed hemolytic transfu-
However, a common error is the overinterpretation sion from AIHA is difficult on occasion.4-7 This differ-
of cold agglutination. Many cold antibodies are reac- ential diagnosis is discussed in Chapter 9.
tive at room temperature but are clinically benign,
albeit somewhat of a nuisance in the laboratory. The
criteria for distinguishing clinically benign cold
Hemoglobinemia and Hemoglobinuria
agglutinins from those pathologic cold agglutinins Hemoglobinuria (hemoglobin in the urine) is far less
capable of causing CAS are discussed later. common than hematuria (RBCs in the urine), and a
very common clinical error is the assumption that a
patient’s red urine is caused by hematuria. It should
Drug Ingestion be remembered that hemoglobinuria, associated with
A temporal history of drug ingestion may suggest the hemolytic anemia, cannot occur without hemoglo-
etiology of the patient’s IHA. A critical aspect of eva- binemia. If red urine is present without hemoglobine-
luation of a patient with IHA is the elicitation of a mia, it should be suspected that the cause is hematuria

FIGURE 5-1. An anticoagulated blood sample from a patient with cold agglutinin syndrome. The cold
agglutinin reacted to a titer of 2048 at 4°C and 512 at 20°C in saline. The figure shows a huge mass
of agglutinated red cells in an anticoagulated blood sample that had been placed at 4°C. Such
agglutination is often mistaken for a clot. However, agglutination completely disperses after incubation
at 37°C for 5 to 10 minutes.
Differential Diagnosis of Immune Hemolytic Anemias 169

and not hemolysis. The presence of hemoglobinemia components, or both. The performance of the DAT
and hemoglobinuria should alert the clinician to a supplies such information. Further tests must be per-
specific group of diagnoses and, when considered in formed to determine the characteristics of the anti-
association with the clinical setting, often makes the bodies in the patient’s serum and in a RBC eluate. In
specific diagnosis evident (see Table 2-6 of Chapter 2). addition to the DAT, we usually perform an “AIHA
The most common associated diagnoses are probably screen,” which is an elaboration of the routine anti-
a hemolytic transfusion reaction and severe acute body screen performed for pretransfusion testing. The
WAIHA, although hemoglobinuria may occur in full technical details of this are given in Chapter 6. In
patients with CAS, especially after exposure to cold. summary, the patient’s serum is tested against a pool
Also, drug-induced HA caused by cefotetan and cef- of untreated and enzyme (e.g., ficin)-treated allo-
triaxone are commonly associated with hemoglobine- geneic group O RBCs and autologous RBCs at 20°C
mia and hemoglobinuria (see Chapter 8). (room temperature can be used) and 37°C (all stages
Hemoglobinuria and hemoglobinemia are much strictly at 37°C). A duplicate set of tubes is used with
more common in children (see Chapter 9). In children, added fresh complement (pooled normal sera) at
both warm and cold AIHA can be the cause. In a child, optimal pH (6.5 to 6.8) for lysis. After incubation, the
PCH should be suspected and a Donath-Landsteiner tubes are inspected for hemolysis, agglutination, and
(DL) test performed whenever an acute severe sensitization (antiglobulin test using polyspecific
hemolytic anemia occurs with hemoglobinemia and antiglobulin serum).
hemoglobinuria. Indeed, hemoglobinuria is a common The results of this screen usually indicate whether
presenting manifestation of PCH3 so that this becomes we are dealing with a cold autoagglutinin, a “warm”
an important diagnostic clue. autoantibody, or possibly a combination of both.
Other points of interest are whether there is a hemo-
Red Blood Cell Morphology and lysin present and whether it is a “cold” or “warm”
hemolysin. If agglutination occurs at 20°C, a cold
Erythrophagocytosis agglutinin titer and thermal amplitude are performed.
One of the easily performed and very valuable tests With the results of the DAT, AIHA screen, and pos-
for determining the specific diagnosis in a patient sibly a cold agglutinin titer/thermal amplitude, we
with hemolytic anemia is examination of the peri- usually have a very good idea of whether we are
pheral blood film. RBC morphology often strongly dealing with WAIHA or CAS. Sometimes the patient’s
suggests a specific diagnosis or a limited number of history and results of the DAT/AIHA screen will lead
diagnostic possibilities (see Chapter 2, page 53). to further tests such as the DL test for PCH, or detec-
Red cell adherence and erythrophagocytosis by tion of drug-dependent antibodies.
neutrophils are prominent findings in PCH but are The full battery of tests described above and in
seen rarely in other forms of immune-mediated Chapter 6 are perhaps best done by specialists (e.g., a
hemolytic anemia. Erythrophagocytosis is rarely reference laboratory), but for most cases, the DAT,
observed in the peripheral blood film of the more antibody screen as performed routinely in pretransfu-
common WAIHA. When it is observed, monocytes, sion testing, together with a cold agglutinin titer, can
not neutrophils, are more often involved (see Chapter yield enough information to support a diagnosis of
3, page 76). WAIHA or CAS. For instance, the addition of comple-
ment (at pH 6.5 to 6.8) is useful only in a small per-
centage of cases (see Tables 5-1 and 5-2).
Association with Mycoplasma
pneumoniae
If a patient with Mycoplasma pneumoniae infection Significance of the DAT in the
develops AIHA, the diagnosis of CAS must be Differential Diagnosis of Immune
strongly suspected because AIHA in this setting is Hemolytic Anemias
almost always caused by cold agglutinins (see The DAT using polyspecific and monospecific
Chapter 3, page 100). antiglobulin reagents provides useful information in
the evaluation of a patient with IHA. However, the
LABORATORY DIAGNOSIS OF IMMUNE results must always be interpreted in conjunction
with clinical and other laboratory data to avoid
HEMOLYTIC ANEMIAS erroneous conclusions. As indicated in Chapter 6, a
positive DAT occurs in situations other than IHAs. A
Even in the presence of valuable clinical clues that positive DAT does not necessarily indicate the
may suggest a specific diagnosis, the confirmation of presence of autoantibody; furthermore, even if auto-
the precise diagnosis of the type of IHA present antibody is present, the patient may or may not have
depends on the laboratory. The serologic tests to be a hemolytic anemia. Thus, an independent assess-
performed determine whether the patient’s RBCs are ment must be made to determine the presence or
coated with immunoglobulin G (IgG), complement absence of hemolytic anemia, and the role of the DAT
170 Immune Hemolytic Anemias

Sensitivity of the test is 96% to 98% because only 2%


TABLE 5-1. RESULTS OF SERUM SCREENING IN to 4% of patients with AIHA have a negative DAT (see
244 PATIENTS WITH WARM ANTIBODY AIHA Chapter 9), but specificity is about 92% to 93% (100%
minus the 7% to 8% incidence). Of 10,000 patients,
Acidified Serum and there would be 10 true positives (TPs) and no false
Serum Acidified Complement
(% Positive Reactions) (% Positive Reactions) negatives (FNs). A total of 690 would be FPs (7% of
9990), leaving 9300 TNs. Thus, the PV of a positive
20°C, Untreated RBCs DAT is 1.4%. Clearly, even in a hospital population,
Lysis 0.4 0.8 the DAT should not be used to screen for AIHA; it
Agglutination 34.8 34.8 would be wasteful to let positive results trigger other
20°C, Enzyme Treated serologic and biochemical tests. Performing a DAT as
Lysis 1.6 2.5 part of a differential diagnosis of hemolytic anemia is
Agglutination 78.6 78.6 another picture entirely (see Table 5-3). If a patient
37°C, Untreated RBCs with hemolytic anemia has a positive DAT, sensitivity
Lysis 0.4 0.4 remains at 98% and specificity at 93%, but disease
Agglutination 4.9 4.9 prevalence changes markedly. We have assumed that
Indirect antiglobulin test 57.4 57.4
one fourth, or 25 of 100, cases of hemolytic anemia
37°C, Enzyme Treated may be autoimmune. If 4% of AIHAs are DAT-
Lysis 8.6 12.7 negative, then there would be 1 FN and 24 TP. Five
Agglutination 88.9 88.9
(7% of 75) would be FP and 71 would be TN. Thus, the
PV of a positive DAT is 83%, which is clearly useful.
The 99% PV of a negative result is also quite impor-
tant in this setting.
is to aid in the evaluation of the etiology of hemolysis Numerous complexities may be considered regard-
when present. ing the DAT, and highly detailed classifications of AIHA
Kaplan and Garratty8 used a predictive value (PV) have been based on DAT results.10-12 However, we
model (Table 5-3) to evaluate the value of the DAT in believe that excessively detailed testing adds little to a
predicting whether a patient has AIHA. The reported clinically significant classification of IHAs. Prognosis
incidence of positive DATs in random hospital and appropriate therapy cannot be correlated with
patients has ranged from approximately 1% up to 15% classifications based on the results of the DAT even
but is usually 7% to 8%, when the antiglobulin reagent when it is performed with a large battery of antisera to
contains optimal anti-C3d activity.9 The prevalence of immunoglobulins and complement components.
AIHA in this group has not been determined, but if
we arbitrarily assign a figure of 1:1000—an almost Results Using Polyspecific and
100-fold increase over that of the general popula- Monospecific Antiglobulin Reagents in
tion—we can calculate the PV of a positive DAT.
Patients with AIHAs
It is convenient to first perform the DAT with a poly-
specific antiglobulin serum, which is defined as one
that must contain anti-IgG and anti-C3d and may
TABLE 5-2. RESULTS OF SERUM SCREENING IN 57 contain antibodies to other complement components
PATIENTS WITH COLD AGGLUTININ SYNDROME (e.g., C3b, C3c, and C4) and to other immunoglobulins
(e.g., IgA and IgM). A positive result in a patient with
Acidified Serum and acquired hemolytic anemia generally indicates that
Serum Acidified Complement the patient’s RBCs are coated with IgG, C3dg, or both.
(% Positive Reactions) (% Positive Reactions) Using monospecific anti-IgG and anti-C3, it is then a
20°C, Untreated RBCs
simple matter to determine which of these two proteins
Lysis 2.0 14.3 are coating the patient’s RBCs. The significance of such
Agglutination 98 98 results is outlined in Tables 5-4 and 5-5.
20°C, Enzyme Treated
Lysis 24.5 93.8
Agglutination 100 100 THE ANTI-COMPLEMENT COMPONENT OF
POLYSPECIFIC ANTIGLOBULIN SERUM
37°C, Untreated RBCs
Lysis 0 0 One point that is evident that must be emphasized is
Agglutination 10.7 10.7
Indirect antiglobulin test 5.4 5.4
that the performance of the DAT with an antiglobulin
serum that does not contain anti-C3d will frequently
37°C, Enzyme Treated result in misleadingly negative results in patients with
Lysis 12.2 22.5
Agglutination 28.6 28.6
IHAs. This is true in all patients with CAS, 13% of
patients with WAIHA, essentially all patients with
Differential Diagnosis of Immune Hemolytic Anemias 171

TABLE 5-3. USING A PREDICTIVE VALUE MODEL TO EVALUATE THE VALUE OF THE DAT IN DIAGNOSING AIHA

Patients with Positive Test Result Patients with Negative Test Result Totals

Patients with disease True positives (TP) False negatives (FN) TP + FN


Patients without disease False positives (FP) True negatives (TN) FP + TN
Totals TP + FP FN + TN TP + FP + TN + FN

Sensitivity: Percent positivity in disease: Specificity: Percent negativity in absence of disease:


TP TN
× 100 × 100
TP + FN FP + FN
Predictive value of a positive result: Predictive value of a negative result:
TP TN
× 100 × 100
TP + FP TN + FN

Screening Random Hospital Patients and Patients with Hemolytic Anemia (HA) for a Diagnosis of AIHA

Positive DAT Negative DAT Total

Random HA Random HA Random HA

AIHA 10 24 0 1 10 25
No AIHA 690 5 9300 70 9990 75
Total 700 29 9300 71 10,000 100
PV of a positive result for random patients = (10/700) × 100 = 1.4%
PV of a positive result for patients with HA = (24/29) × 100 = 83%
[PV of a negative test = (70/71) × 100 = 99%]

PCH, and many instances of drug-induced IHAs. tion of the RBCs4 (see Table 5-5). Other reported series
Altogether 26% of the patients in our series had a of patients with AIHA yielded comparable informa-
positive DAT caused only by complement sensitiza- tion. For instance, Dacie and Worlledge13 reported
that 33% of 29 patients with AIHA had RBCs sensi-
tized only with complement components.
In regard to antibodies against other complement
TABLE 5-4. TYPICAL DIRECT ANTIGLOBULIN components in antiglobulin serum, we have occasion-
TEST RESULTS IN PATIENTS WITH IMMUNE ally found patients whose RBCs are sensitized with
HEMOLYTIC ANEMIAS C4d but not C3d. However, none of these patients has
had hemolytic anemia.14,15 Patients with IHA fre-
Type of Immune Hemolytic Anemia Anti-IgG Anti-C3 quently have C4d on their RBCs in addition to C3d,
but in testing hundreds of patients with AIHA, we
Autoimmune Hemolytic Anemias
Warm antibody AIHA
have not observed a single patient with hemolysis
67% + + whose RBCs had C4d but not C3d. We know of no
20% + 0 experience contrary to this in the medical literature.
13% 0 +
Warm antibody AIHA associated with
systemic lupus erythematosus + +
Cold agglutinin syndrome (100%)* 0 +
Paroxysmal cold hemoglobinuria (100%) 0 +
TABLE 5-5. RESULTS OF DIRECT ANTIGLOBULIN
Drug-induced Immune Hemolytic Anemias TEST WITH ANTI-IgG AND ANTI-C3 IN 347
Drug-dependent antibodies
Penicillin and first-generation cephalosporins + (+) PATIENTS WITH AIHA AND DRUG-INDUCED
Second- and third-generation cephalosporins + + IMMUNE HEMOLYTIC ANEMIA
Associated with “immune complex mechanism” (+) +
Drug-independent antibodies + (+) Percentage*
Drug-induced nonimmunologic adsorption of
proteins + (+) IgG (no C3) 23
Hemolytic Disease of Fetus/Newborn + 0 73% have IgG on RBC
Hemolytic Transfusion Reactions + (+) IgG + C3 50
76.4% have C3 on RBC
(+) = Sometimes positive C3 (no IgG) 26.4
* We detected IgG +C3 on the RBCs of one patient with CAS because the
patient was on methyldopa therapy and made IgG autoantibodies. * Two patients (0.6%) had only IgA present on their RBCs.
172 Immune Hemolytic Anemias

Therefore, anti-C4 appears to be superfluous in serum, ELISA, and flow cytometry26-32 have largely
antiglobulin serum used for detection and differential replaced the use of antiglobulin test titrations and
diagnosis of IHA. Similarly, although with less exten- scores. Nevertheless, some useful information has
sive data available, antibodies to other complement been gained using this technique in the past.
components in antiglobulin serum (C5, C6, C8) have For example, clinical remission of AIHA is fre-
proven to be of little significant clinical value.14,16,17 quently associated with a significant decrease in the
One exception was a single report suggesting that C5, strength of the DAT, although this may not be evident
without the presence of C3d, may sometimes be on if one uses only a single dilution of the antiglobulin
the RBCs of rare patients with AIHA.18 serum. It should be noted that these dilutions were
performed using raw antiglobulin sera, not commer-
cial products, which are already diluted optimally for
SIGNIFICANCE OF ANTI-IgM AND ANTI-IgA routine use. The 1:128 dilution in Table 5-6 is probably
IN ANTIGLOBULIN SERUM close to undiluted commercial anti-IgG. Table 5-6
illustrates the results of testing a patient’s RBCs with
Even with potent antisera, RBC-bound IgM is difficult dilutions of anti-IgG during relapse and during clini-
to detect with the antiglobulin test.4,19,20 Fortunately, cal remission. The decrease in strength of sensitization
IgM antibodies that cause IHA characteristically fix of the RBCs is not obvious with the 1:128 dilution (or
complement that is much more readily detected. undiluted commercial anti-IgG) but is obvious after
AIHA associated with warm IgM autoantibodies using a series of dilutions of the anti-IgG. Chaplin33,34
without the presence of IgG occurs on rare occasions has reported similar observations. The fact that the
and is discussed later in this chapter and in Chapter 9. DAT is often markedly reduced in strength during
Anti-IgA and anti-IgM that are standardized for the remission is frequently missed because DAT titrations
DAT are not readily available in the United States. are now rarely performed.
Nevertheless, one can standardize immunologic There have been published studies on the relation-
reagents for use in the DAT to detect for detection of ship between antiglobulin test titration scores and
IgA and IgM (see Chapter 6, page 210). These reagents quantitative measurements of RBC sensitization. We
are of significance in evaluating exceptional patients used an immunochemical method to quantitate the
with AIHA, particularly those whose AIHA is associ- number of C3 molecules bound to human RBCs in
ated only with IgA autoantibodies or warm IgM vitro or in vivo, and then compared these results with
autoantibodies. IgM sensitization of RBCs is more the antiglobulin test and the antiglobulin test titration
readily detected using flow cytometry than by the scores.35 As illustrated in Figures 5-2 and 5-3, a wide
DAT21-24; flow cytometry is also useful for detecting range of cell-bound C3 molecules may result in a 2+ to
RBC-bound IgA.25 4+ DAT using a single dilution of antiglobulin serum.
IgA antibodies only infrequently play a role in RBC In contrast are the data illustrated in Figures 5-4 and
sensitization, and in such cases, other immune glo- 5-5, which indicate that antiglobulin test scores corre-
bulins and/or complement components are almost lated quite well with the immunochemical assessment
always, although not invariably, found on the cell of the number of molecules per RBC. There is also a
surface as well. Occasionally patients with WAIHA good correlation between the antiglobulin test titra-
have a DAT that is positive only with anti-IgA. The tion scores and quantitative measurements of RBC
clinical and hematologic features of WAIHA associ- bound IgG with RBCs sensitized with fewer than 1000
ated only with IgA autoantibodies are very similar to molecules of IgG per RBC as is illustrated in Figure 5-6.
AIHA associated with warm IgG autoantibodies. Fischer and colleagues35 further studied 25 patients
Because WAIHA associated with IgA autoantibodies who had positive DATs that were caused at least in
usually presents as AIHA with a negative DAT, such
cases are discussed in Chapter 9.

TABLE 5-6. ANTIGLOBULIN TEST TITRATIONS


Antiglobulin Test Titrations and Scores IN A PATIENT WITH WAIHA IN RELAPSE AFTER
The amount of protein sensitizing RBCs can be judged REMISSION INDUCED BY CORTICOSTEROID
in a semiquantitative way by simply testing the RBCs THERAPY
with dilutions of antiglobulin serum. The strength of
Reciprocals of
agglutination at each dilution of the antiglobulin Dilutions of
serum can then be used to develop an antiglobulin Our Anti-IgG* 128 256 512 1024 2048 4096 Score
test titration score. For example, 4+ agglutination may
be assigned a value of 10; 3+ agglutination, a value of Relapse 4+ 4+ 4+ 3+ 2+ 1+ 48
8; 2+, a value of 6; 1+, a value of 4; and 1⁄2+, a value Remission 4+ 2+ 1+ 0 0 0 18
of 2. More accurate ways of measuring the amount of Agglutination reactions are graded as 1⁄2+ to 4+.
RBC-bound proteins using radiolabeled antiglobulin * Raw (undiluted) “homemade” anti-IgG (see text above).
Differential Diagnosis of Immune Hemolytic Anemias 173

23,850
12,000 13,650 16,650

5.0

9600
4.5
Number of C3 molecules/RBC

Log of number of C3 molecules/RBC


4.0
7200
3.5

4800 3.0

2.5

2400
2.0
1200
1.5
1/2 1 11/2 2 3 4
Antiglobulin test 1.0
3 7 11 15 19 23 27 31 35
FIGURE 5-2. The relationship between the number of C3 molecules Antiglobulin test scores
per red cell and the antiglobulin test using a single dilution of anti-C3
antiserum. C3-sensitized RBCs were prepared in vitro using Lea FIGURE 5-4. Correlation between antiglobulin titration scores and the
antibody in varying dilutions. (From Fischer JT, Petz LD, Garratty G, quantitative assay of cell-bound C3 (in vitro sensitized RBCs). The linear
Cooper NR: Correlations between quantitative assay of red-cell bound regression coefficient with 95 percent confidence limits is 0.071 ±
C3, serologic reactions, and hemolytic anemia. Blood 1974;44:359.) 0.005. The coefficient of correlation by linear regression (r) = 0.83,
significant at the 1 percent level. (From Fischer JT, Petz LD, Garratty G,
Cooper NR: Correlations between quantitative assay of red-cell bound
C3, serologic reactions, and hemolytic anemia. Blood 1974;44:359.)

7500
2400

4.5
2000
Log of number of C3 molecules/RBC

4.0
Number of C3 molecules/RBC

1600
3.5

1200 3.0

2.5
800
2.0

400
1.5

1.0
1/2 1 2 3 4 3 7 11 15 19 23 27 31 35
Antiglobulin test Antiglobulin test scores
FIGURE 5-3. The relationship between the number of C3 molecules FIGURE 5-5. Correlation between DAT titration scores and the
per RBC and the antiglobulin test using a single dilution of anti-C3. quantitative assay of cell bound C3 (RBCs sensitized in vivo). The linear
C3-sensitized RBCs were obtained from patients having a positive DAT regression coefficient with 95 percent confidence limits is 0.031 ±
caused at least in part by RBC sensitization by C3. (From Fischer JT, 0.012. The coefficient of correlation by linear regression (r) = 0.66,
Petz LD, Garratty G, Cooper NR: Correlations between quantitative significant at the 1 percent level. (From Fischer JT, Petz LD, Garratty G,
assay of red-cell bound C3, serologic reactions, and hemolytic anemia. Cooper NR: Correlations between quantitative assay of red-cell bound
Blood 1974;44:359.) C3, serologic reactions, and hemolytic anemia. Blood 1974;44:359.)
174 Immune Hemolytic Anemias

IgG; and 2% had only IgA. Sokol and coworkers39


found only 24% of their AIHA patients to have RBC-
bound IgG+ C3 and 61% had IgG alone. These differ-
ences might be due to the amount of anti-C3dg in the
anti-C3 used for the DAT. Other patterns may also be
found if additional antiglobulin sera are used (e.g.,
anti-IgA and -IgM)4 (Table 5-9).
Hsu and coworkers19 studied 34 patients with posi-
tive DATs using an AutoAnalyzer, which was more
sensitive than the routine DAT. None of the 16 patients
without hemolytic anemia had RBC-bound C3 or IgM;
88% had RBC-bound IgG; and 25% had RBC-bound
IgA (2 patients had only IgA detected on the RBCs).
RBC-bound IgG was detected on the RBCs of 11 of 13
patients (85%) with AIHA; 61% had RBC-bound C3;
FIGURE 5-6. Correlation between antiglobulin test titration scores and 69% had RBC-bound IgA; and 61% had RBC-bound
a quantitative immunochemical assay of red cell bound IgG. IgM. The authors emphasized that hemolytic anemia
appeared to be associated commonly with RBC-bound
C3, IgA, and IgM in addition to IgG, in contrast to the
patients without hemolytic anemia. They noted that
part by RBC-bound C3. Only 2 of 14 patients with less RBC-bound IgM was not detected when the hemolytic
than 1100 molecules of C3 per RBC had hemolytic anemia was in partial or complete remission. It should
anemia, whereas 8 of 11 patients with at least 1100 be emphasized that the AutoAnalyzer may detect RBC-
molecules of C3 per RBC did have overt hemolysis. bound proteins not detectable by the routine DAT, even
The presence or absence of hemolysis was not when anti-IgM is used. It is interesting to note that
explained by variations in the amount of IgG on these Lalezari and coworkers40 found that the RBCs from
patients’ RBCs as assessed by the antiglobulin test patients with hemolytic anemia due to methyldopa
using monospecific anti-IgG (Table 5-6). therapy had RBC-bound IgM (and C1q) in addition to
Garratty and Nance29 showed that flow cytometry IgG. When the hemolysis stopped, no RBC-bound IgG
was more efficient at differentiating the degree of RBC was detectable. Patients with positive DATs but no
sensitization with IgG than test titration scores, when hemolytic anemia did not have RBC-bound IgM. The
RBCs were strongly sensitized (e.g., 4+ antiglobulin RBC-bound IgM was only detectable using an Auto-
tests). Table 5-8 shows the results. Analyzer. It was suggested that the RBC-bound IgM
The significance of semiquantitative or quantitative was monomeric. No one has confirmed this interesting
measurements of the amount of IgG or C3 on the RBC finding.
membrane must not be exaggerated. Rosse36,37 and Ben-Izhak and coworkers41 suggested that the pres-
Garratty and Nance29 demonstrated that the amount ence of more than one class of immunoglobulin on
of antibody detected on the RBC membrane was RBCs, detectable by the standard DAT, was associated
generally proportional to the rate of RBC destruction, with severe hemolytic anemia. They found that 36% of
but exceptions to this correlation were frequent. 25 patients with AIHA had IgM or IgA in addition to
Indeed, in an individual patient, it is impossible to IgG on their RBCs. These patients appeared to respond
predict the presence or absence of a shortened RBC less well to steroid therapy. Using an enzyme-linked
life span on the basis of measures of RBC bound antiglobulin test (ELAT), Sokol and coworkers42
immunoglobulins. The in vivo significance of a given showed that multiple immunoglobulins could be
degree of RBC sensitization by autoantibodies varies detected on 35% of 153 patients who were suspected of
greatly, even among antibodies of the same immu- having AIHA. Only 7.2% showed the same results
noglobulin class.29 when the standard DAT (anti-IgG+ anti-C3) was used.
Using ELAT, the following immunoglobulins were
Warm Antibody AIHA detected: 23% of the RBCs were sensitized with IgG and
IgM; 5.8% were sensitized with IgG plus IgA; and 8%
In WAIHA, there is no diagnostic pattern of DAT reac- were sensitized with IgG, IgM, and IgA. RBC-bound
tivity. Most commonly, positive reactions are obtained IgG was present in larger amounts than IgM or IgA in
both with anti-IgG and anti-C3, but in a minority of 63% of the samples. Compared with IgG sensitization
cases, only IgG or C3 is found on the patient’s RBCs. alone, multiple immunoglobulins were significantly
Using these antisera to test the RBCs of 244 patients associated with larger quantities (>800 molecules/RBC)
with WAIHA, we found 67% had RBC-bound IgG and of IgG, multiple IgG subclasses, IgG+C3d bound to
C3; 20% had RBC-bound IgG but no C3; and 13% had RBCs, and with serum haptoglobin levels of less than
RBC-bound C3 but no IgG10 (see Table 5-4). Worlledge 0.1 g/L. The latter association was still significant when
and Blajchman38 found that 48% had RBC-bound higher levels of RBC-bound IgG and IgG subclass
IgG+ C3; 39% had IgG but no C3; 11% had C3 but no pattern were taken into account. In RBC samples
TABLE 5-7. RESULTS OF ASSAYS OF RBC-BOUND C3 IN 25 PATIENTS AND CORRELATION WITH CLINICAL AND SEROLOGICAL DATA

No. of C3 DAT (Anti-C3) Hemolytic


Disease DAT (Anti-C3) Molecules per RBC Titration Score Anemia Hct Hgb Reticulocytes (%) DAT (Anti-IgG)

1. SLE 1⁄ +
2 60 1 No 41.7 14.8 — 1+
2. Iron deficiency anemia 1⁄ +
2 90 1 No 21.1 7.0 3 0
3. Normal 1+ 180 2 No 39.8 14.0 — 0
4. SLE 1+ 235 8 Yes 30.2 10.4 4.8 1+
5. Hypertension 2+ 410 7 No 44.4 15.8 — 3+
6. DIIHAa (Aldomet) 1+ 465 2 Yes 37.5 12.6 3.1 4+
7. Normal 1+ 625 2 No 42.0 — — 0
8. SLE 1+ 630 3 No 41.5 13.4 — 0
9. Vasculitis 2+ 700 16 No 40.0 14.1 2 0
10. SLE 2+ 720 18 No 44.0 14.7 — 2+
11. SLE 2+ 735 15 No 42.2 14.3 — 2+
12. SLE 2+ 1010 8 No 41.7 14.8 — 0
13. WAIHA (post-splenectomy) 2+ 1050 6 No 42.0 13.2 — 2+
14. Chronic active hepatitis 2+ 1075 15 No 40.1 13.3 — 2+
15. DIIHAa (penicillin) 2+ 1100 16 Yes 11.0 3.6 26 2+
16. Acute lymphocytic leukemia 2+ 1125 11 Yes 17.0 4.5 0.3b 3+
17. PCH 3+ 1260 21 Yes 35.0 11.2 6.3 0
18. SLE 3+ 1335 30 No 26.7 8.5 2.1 1+
19. WAIHA 3+ 1340 31 Yes 31.0 9.3 10.2 0
20. Rheumatoid arthritis 3+ 1510 15 Noc 34.0 10.0 2.0 3+
21. SLE 3+ 1560 21 No 38.0 12.9 0.8 4+
22. WAIHA 2+ 1710 16 Yes 33.0 10.7 1.5 1+
23. CAS 4+ 1910 33 Yes 24.0 7.8 9.2 0
24. CAS 3+ 2375 19 Yes 20.5 7.1 4.4 0
25. CAS 4+ 7500 35 Yes 22.4 7.8 11.6 0
a DIIHA, drug-induced immune hemolytic anemia.
b Hemolysis was present as indicated by an excessive transfusion requirement without evidence of blood loss. The patient’s low reticulocyte response was presumably related to his acute leukemia.
c Later this patient developed overt hemolytic anemia, with the hematocrit level falling to as low as 10 percent and the reticulocytes increasing to a maximum of 30%.
Data from Fischer JT, Petz LD, Garratty G, Cooper NR: Correlations between quantitative assay of red cell-bound C3, serologic reactions, and hemolytic anemia. Blood 1974;44:359–373.

175
176 Immune Hemolytic Anemias

RBC) of IgG, multiple IgG subclasses, IgG3 and C3d


TABLE 5-8. ANTIGLOBULIN TEST TITRATION bound to the cells, and with serum haptoglobin levels
SCORES AND FLOW CYTOMETRIC ANALYSIS of less than 0.1 g/L. The latter association was still
OF RBCs SENSITIZED IN VITRO WITH significant when higher levels of RBC-bound IgG and
DILUTIONS OF ANTI-D subclass pattern were taken into account. It was con-
cluded that multiple immunoglobulin coating, even
Indirect when undetected by agglutination methods, is a
Anti-D Antiglobulin Titration Fluorescence Percentage of
Dilution Test Score per RBC Fluorescence major cause of hemolysis; it is part of a more general-
ized autoimmune response and acts with other factors
1:5 4+ 68 212.0 100 such as the quantity of bound IgG, the IgG subclass
1:10 4+ 68 183.2 100 pattern, and complement; it also has an important
1:20 4+ 62 130.1 100 hemolytic effect in its own right.
1:40 4+ 65 84.7 99.4

Data from Garratty G, Nance SJ: Correlation between in vivo hemolysis and
the amount of red cell-bound IgG measured by flow cytometry. Transfusion Cold Agglutinin Syndrome
1990;30:617–621.
In contrast to the variable findings in WAIHA, the
DAT in CAS is invariably positive with anti-C3.
Equally important is the fact that reactions are almost
with multiple immunoglobulin sensitization, there was always negative with anti-IgG; that is, a positive DAT
no significant relationship (p > 0.05) between haptoglo- with anti-IgG essentially excludes CAS as the sole
bins of less than 0.1 g/L and RBC-bound C3d or multi- diagnosis (see Table 5-4).
ple IgG subclasses. The authors concluded that RBC
sensitization with multiple immunoglobulin classes, Paroxysmal Cold Hemoglobinuria
even when undetected by the DAT, may be an impor-
tant factor in determining the severity of AIHA. PCH is caused by an IgG complement fixing antibody
Sokol and coworkers43 extended the above in 404 but, nevertheless, the DAT is usually positive only
patients with warm-reactive RBC autoantibodies on with anti-C3 (see Table 5-4). If RBCs are washed at
590 occasions. Multiple immunoglobulins were 37°C, no RBC-bound IgG is detected. Negative reac-
detected by enzyme-linked DATs in 218 samples tions with anti-IgG are probably caused by the fact
(37%), but in only 87 (15%) by DAT (i.e., by agglutina- that the DL antibody readily elutes from the RBC
tion). RBC-bound C3d was detected on the RBCs of membrane during in vitro washing, whereas comple-
64% of the patients. Compared with IgG coating ment components remain fixed to the cell membrane.
alone, multiple immunoglobulins were significantly Dacie and Worlledge13 reported that if the patient’s
associated with larger quantities (>800 molecules/ RBCs are washed in saline at 10°C or below, the DAT
may be positive using anti-IgG antiserum as well as
anti-C3. Gottsche and coworkers44 reported that the
DAT was positive using anti-IgG in 6 of 22 patients
(27%) if the RBCs were washed at 20°C but invariably
TABLE 5-9. DAT RESULTS IN 104 PATIENTS negative if RBCs were washed at 37°C. The DAT will
WITH WAIHA USING SEVERAL MONOSPECIFIC be positive at the time of a paroxysm of hemoglobin-
ANTIGLOBULIN SERA uria and for a variable duration of time thereafter.13,44

Percentage
Drug-Induced Immune Hemolytic Anemia
IgG only 18.3 DAT results in patients with drug-induced IHAs varies
C3 only 10.6
IgA only 1.9 and generally correlates with the mechanism of devel-
IgM only 0* opment of the drug-induced antibodies (see Chapter 8).
IgG and C3 46.2 Typically, when penicillin or first-generation cephalo-
IgG and C3 and IgA 12.5 sporins cause IHA by the “drug-adsorption mecha-
IgG and C3 and IgA and IgM 1.9
IgG and IgA 2.9
nism,” the DAT is positive with anti-IgG; a minority of
IgG and IgM 0* patients may have RBC-bound complement detected as
IgG and IgM and C3 3.9 well. The DATs associated with IHA due to second- and
IgG and IgA and IgM 0* third-generation cephalosporins are usually strongly
C3 and IgA 1.9 positive due to IgG plus C3 or, less commonly, C3 with
C3 and IgM 1.9
IgG present alone or together with other proteins 85.6 no IgG sensitization.45 Drugs causing IHA via the
C3 present alone or together with other proteins 78.9 “immune complex mechanism” characteristically
IgA present alone or together with other proteins 21.2 result in the DAT being positive with anti-C3, but in a
IgM present alone or together with other proteins 7.7 minority of instances RBC-bound IgG and/or IgM may
* Since studying these 104 patients, we have encountered rare patients who also be present. Patients with drug-induced AIHA
have IgM but no C3 on their RBCs. usually have positive DATs associated with IgG
Differential Diagnosis of Immune Hemolytic Anemias 177

sensitization; a few patients may also have weak C3 If a patient has an acquired hemolytic anemia with a
sensitization. Drug-induced nonimmunologic adsorp- positive DAT but no RBC antibodies are demonstrable
tion of proteins may cause the DAT to be positive as a in either the serum or RBC eluate, such findings lend
result of a multitude of proteins on the RBC surface weight to a diagnosis of a drug-induced hemolytic
(e.g., IgG, IgM, IgA, C3, and albumin). anernia. This is true because drug-dependent antibo-
dies will give negative results in serologic studies
unless the offending drug is in the in vitro test system.
Systemic Lupus Erythematosus However, one must also keep in mind that drug-
Systemic lupus erythematosus (SLE) deserves parti- induced IHAs are very uncommon, whereas positive
cular consideration, because many patients with this DATs that yield nonreactive eluates are quite common.
disorder have C3 on their RBCs even when no evi- Indeed, up to 80% of all positive DATs yield nonreac-
dence of hemolysis exists. Budman and Steinberg46 tive eluates as occurs when the DAT is due to nonim-
reported that the DAT was positive in 18% to 65% of munologic uptake of IgG onto the patient’s RBCs.
patients in various published series of patients with Alloantibody-induced IHAs (hemolytic disease of
SLE but only about 10% of these patients had clini- the newborn and acute hemolytic transfusion reac-
cally significant hemolysis. When the DAT was posi- tions) are usually made evident by the clinical setting.
tive in a patient whose SLE was quiescent, it was However, a delayed hemolytic transfusion reaction
positive with anti-C3 but negative with anti-IgG; in may not be obvious because the hemolysis may
patients with overt hemolysis, the DAT was more develop at least several days following the transfusion
commonly positive with both anti-C3 and anti-IgG as (see Chapter 9).
indicated in Table 5-4. Rarely, the DAT was positive Thus, depite the lengthy list of diagnostic possibili-
only with anti-IgG. Mongan and coworkers47 reported ties, it is usually optimal to perform the initial sero-
similar findings. logic tests with only three diagnostic considerations in
mind: WAIHA, CAS, and PCH.
If findings are not characteristic of any of these
An Approach to the Characterization of diagnoses, the patient may have a drug-induced IHA
Antibodies in the Serum and Eluates (Chapter 8), or an atypical hemolytic anemia, such as
from RBCs of Patients with AIHA AIHA with a negative DAT (Chapter 9), AIHA caused
by IgM warm autoantibodies (see later, page 180),
Although the DAT provides useful information, the AIHA associated with IgA autantibodies (Chapter 9),
definitive diagnosis rests on the characterization of or AIHA caused by IgG cold autoantibodies (see later,
the antibodies present in the patient’s serum and in an page 190). Collectively, such cases constitute only a
eluate from the patient’s RBCs. It is wise to initiate small percentage of cases of IHAs, but nevertheless
such studies with screening tests to develop a prelim- are an important group of patients.
inary diagnosis and then to perform additional tests Using the “AIHA Screen” as an Aid in the
as are necessary for confirmation of the diagnosis and Differential Diagnosis of AIHA. Although a routine
for the exclusion of alternative possibilities. blood bank antibody screen, together with a cold
Although the differential diagnosis of IHAs appears agglutinin titer, can sometimes provide enough infor-
complex (see Table 3-1), the initial evaluation can mation to help in the differential diagnosis of AIHA, we
often be simplified in concept by keeping the follow- find more extended testing is necessary to be certain of
ing points in mind. the diagnosis. We have found the “AIHA screen,”
The differentiation between idiopathic and second- described in detail in Chapter 6 and in summary on
ary AIHA is not based on serologic tests but instead page 169 of this chapter, to be immensely helpful in
concerns the usual tests pertinent to the diagnosis of classifying difficult cases.
SLE, chronic lymphocytic leukemia, lymphomas, etc. Tables 5-1 and 5-2 show the results we obtained in
Drug-induced IHA may in some cases be excluded the sera from 244 patients with WAIHA and 54
on the basis of the patient’s history. However, histories patients with CAS, respectively. Sera from patients
of drug administration are notoriously inaccurate (see with PCH often show negative test results at all
Chapter 8). For example, if the drug was given during phases of the AIHA screen (see later). Thus, although
surgery, the patient may not be aware of having there are obvious differences in the AIHA screen
received it and its administration may be indicated results, there is also overlap between WAIHA and
only in obscure locations (e.g., anesthesiologists notes) CAS, indicating that other data (e.g., DAT and cold
in the medical record. Also, with some drugs (e.g., agglutinin titer/thermal amplitude) are necessary to
second- and third-generation cephalosporins), the finalize the diagnosis. The AIHA screen has been very
hemolytic anemia may not develop for a week or useful in helping classify unusual cases. For instance,
more following drug administration. If drug-induced the finding of a hemolysin at 37°C, but not 20°C, may
IHA is a plausible diagnosis, additional studies are help classify a patient as WAIHA when other results
often necessary to demonstrate the drug-related red are suggestive of CAS (e.g., complement but no IgG
cell antibody. These tests are discussed separately in on patient’s RBCs, and a 4+ cold autoagglutinin at
Chapter 8. room temperature). On the other hand, a hemolysin at
178 Immune Hemolytic Anemias

20°C, in the presence of a similar DAT and agglutina- –55°C


100
tion patterns, may be more suggestive of CAS, which

Percentage of complement
– 20°C
would be confirmed by a high thermal amplitude 80
(≥30°C) agglutinin.
60
Characteristic Serology of WAIHA
40
Autoantibodies causing WAIHA are usually IgG but 37°C R.T. 4°C
can be IgM or IgA. As discussed on pages 180–182, 20
these proteins can be present together; it is rare for
only IgM or IgA to be the cause of AIHA (see Table
0
5-9). Table 5-1 shows the characteristics of the anti- 24 48 72 1 2 3 4 8 12
bodies associated with 244 cases of WAIHA. Because Hours Weeks
the patient’s RBCs are adsorbing warm autoantibod-
ies continuously, autoantibodies are usually only FIGURE 5-7. Average complement activity of 12 normal sera as
found in the serum when all autoantigens are satu- measured by antiglobulin test assay after storage at various
temperatures. (From Garratty G: The effects of storage and heparin
rated. Only about 60% of patients’ sera will react with on the activity of serum complement, with particular reference to the
saline-suspended RBCs, but a higher percentage will detection of blood group antibodies. Amer Clin Pathol 1970;54:531.)
react in the presence of potentiators (e.g., polyethy-
lene glycol) or enzyme-treated RBCs (Table 5-1).
Some sera contain warm hemolysins, which react IgG SUBCLASS OF RBC-BOUND IgG
optimally at 37°C but may react at 20°C. The hemoly-
sis is enhanced by adding fresh complement at pH 6.5 Nance and Garratty49 and Garratty50 reported the IgG
to 6.8 to the patients’ sera. Von dem Borne and subclass of the autoantibody on RBCs of 167 indi-
coworkers48 showed that such hemolysins were viduals with positive IATs (46 patients with AIHA,
usually IgM autoantibodies; they were maximally and 32 patients and 89 blood donors with a positive
reactive at a pH of 6.5, and 7 of 11 (64%) reacted opti- DAT but no hemolytic anemia). Table 5-11 shows the
mally at 30°C; 4 of 11 reacted optimally at 37°C.48 We results. In all three groups the RBCs were sensitized
found that about one third of sera from patients with with predominantly IgG1 autoantibodies (89%, 85%,
WAIHA contain IgM cold autoagglutinins that can and 87%, respectively); usually IgG1 was the only
react quite strongly at 20°C (or room temperature) but subclass present. IgG2 was present alone, or together
have a normal cold agglutinin titer at 4°C and do not with other subclasses, on the RBCs of 5.8% of donors,
react at 30°C. We do not think that these are patho- and 15.6% and 26.2%, respectively, of the RBCs from
genic but may be naturally occurring cold antibodies patients, without and with AIHA. IgG2 was not found
that become boosted (e.g., raised thermal amplitude) often as the only subclass on the RBCs, but it is inter-
during the pathogenic autoimmune response. esting to note that one patient with hemolytic anemia
A diagnosis can usually be reached on the basis of had only IgG2 detected on the RBCs. IgG3 was
the serologic tests described thus far. Despite the detected on the RBCs of 7% of donors, and 13% and
seemingly complicated nature of the foregoing, the 29%, respectively, of the RBCs of patients without and
usual or “typical” essential diagnostic tests that lead with AIHA; similar to IgG2, it was unusual to find
to a reasonably confident diagnosis of WAIHA may be IgG3 alone on the RBCs. IgG4 was detected on the
very simply summarized as follows: (a) the presence RBCs of 12% of donors, and 13% and 24% of patients
of an acquired hemolytic anemia, (b) a positive DAT, without and with AIHA, respectively, of the RBCs; it
and (c) an unexpected antibody in the serum and was detected as the sole sensitizing subclass in only
eluate that reacts optimally at 37°C. The antibody five cases, and all were blood donors. Dacie51 found a
usually reacts with all normal RBCs but, in some similar distribution of IgG subclasses on the RBCs of
cases, it can readily be shown to react preferentially 57 patients with AIHA. Most of our results were
with antigens on the patient’s own RBCs. similar to those reported by Engelfriet and cowork-
Examples of typical serologic findings in a patient ers52 except for our one unusual case of IgG2-associ-
with WAIHA are listed in Table 5-10. ated AIHA, and the cases with IgG3 sensitization
When performing tests for lysis in vitro, one must associated with no obvious hemolytic anemia.
keep in mind that the patient’s serum is frequently Engelfriet and coworkers52 found that IgG3 sensitiza-
deficient in complement, so it is advisable to add fresh tion was always associated with overt hemolysis. As
complement. This is conveniently done by adding an can be seen in Table 5-11, although IgG3 sensitization
equal volume of ABO compatible normal serum, which was far more common (29%) in patients with
has been shown to contain no unexpected antibodies, to hemolytic anemia than in donors or patients without
the patient’s serum. Figure 5-7 shows the effects of dif- AIHA, we found six (7%) DAT-positive blood donors
ferent storage conditions on serum complement levels. and four (13%) patients with IgG3 RBC sensitization,
It is logical and convenient to perform tests for but no obvious signs of hemolytic anemia. Engelfriet
specificity of the autoantibody using the screening test and coworkers53 reported on the subclass of RBC-
that gave the strongest reactions. Usually this test is bound IgG of 746 patients with a positive DAT. The
the IAT or agglutination of enzyme-treated RBCs. patients were not categorized into those with and
Differential Diagnosis of Immune Hemolytic Anemias 179

TABLE 5-10. SEROLOGIC FINDINGS IN A TYPICAL PATIENT WITH WARM ANTIBODY AUTOIMMUNE
HEMOLYTIC ANEMIA

Screening Tests for Serum Antibody

Controls

Serum AS + AC AC Alone Auto*

20°C, Untreated Cells


Lysis 0 0 0 0
Agglutination 0 0 0 0
20°C, Papainized Cells
Lysis 0 0 0 0
Agglutination 2+ 2+ 0 2+

Serum Controls

Saline Alb AS + AC AC Alone Auto* with Alb

37°C, Untreated Cells


Lysis 0 0 0 0 0
Agglutination 0 1+ 0 0 1+
IAT 1+ 3+ 1+ 0 3+
37°C, Papainized Cells
Lysis 0 NT 0 0 0
Agglutination 3+ NT 2+ 0 3+

Specificity of Antibody in Serum and Eluate


Serum: no obvious specificity (all routine panel RBCs reacting 1+ to 2+)
Eluate: no obvious specificity (all routine panel RBCs reacting 3+)
Cold agglutinin titer: not performed (no agglutination in screening tests at 20°C)
Other data:all serum autoantibody removed by warm autoadsorptions (see Chapter 10)
Summary: findings typical of WAIHA
Follow-up: rapid improvement of anemia over two week period of prednisone therapy

AS, acidified serum; AC, acidified complement; Alb, albumin; NT, not tested
* Auto, patient’s own RBCs.

those without hemolytic anemia. Ninety-six percent Nance and Garratty49 reported that eluates made
had IgG1 on their RBCs, 12% had IgG2, 13% had IgG3, from DAT+ RBCs often contained extra subclasses not
and 3% had IgG4 on their RBCs. Although 74% had detected by the DAT using anti-IgG1, -IgG2, -IgG3, and
only IgG1 on their RBCs, IgG2, IgG3, and IgG4 were -IgG4. They also found that generally, the stronger the
usually present with other subclasses (0.7% had only DAT, the greater was the incidence of multiple IgG sub-
IgG2, 2% had only IgG3, and 0.9% had only IgG4 on classes and the degree of hemolytic anemia; 39% of
their RBCs).53 patients with AIHA had multiple subclasses detected
on their RBCs compared with only 13 of patients/
donors without hemolytic anemia. All these findings
TABLE 5-11. IgG SUBCLASS OF AUTOANTIBODIES were confirmed by Sokol and coworkers.54 Sokol and
ON DAT+ RBCs OF BLOOD DONORS AND PATIENTS coworkers54 used an ELAT to subclass the IgG eluted
WITH AND WITHOUT AIHA from the RBCs of 304 patients on 426 occasions. IgG1
was most common, being found in 98% of cases and as
Blood Patients (%) the sole subclass in 64%; multiple subclasses occurred in
Donors (%) No AIHA AIHA 34.5%. The IgG subclass pattern possibly changed with
IgG Subclass (n = 46) (n = 89) (n = 32) time (p < 0.02, > 0.01), populations being compared at 6
and 12 months. There was a highly significant and
IgG1 alone 81 72 50
IgG2 alone 1.3 6.3 2.2
important correlation between multiple IgG subclasses
IgG3 alone 2.6 6.3 6.6 and multiple immunoglobulin coating; in our further
IgG4 alone 6.5 0 0 studies, this necessitated the use of samples where only
IgG1 with other subclasses 7.9 12.5 37 cell-bound IgG was increased. Multiple IgG subclasses
IgG2 with other subclasses 4.5 9.4 24 strongly correlated with larger amounts of cell-bound
IgG3 with other subclasses 4.5 6.3 22
IgG4 with other subclasses 5.6 12.6 24 IgG, groups with more than 2 and less than 1 OD unit
by the ELAT being compared (approximately more than
Data from Garratty G: Autoimmune hemolytic anemia. In Garratty G (ed): 800 and less than 400 molecules IgG per red cell, respec-
Immunobiology of transfusion medicine. New York: Dekker, 1994:493–521;
and Nance S, Garratty G: Subclass of IgG on red cells of donors and patients tively). Multiple subclasses (p < 0.05, > 0.01), but not
with positive direct antiglobulin tests (abstr). Transfusion 1983;23:413. IgG3, were possibly associated with low haptoglobin
180 Immune Hemolytic Anemias

levels; significance was reached, however, if the multi- and/or complement is detectable by the antiglobulin
ple immunoglobulin effect was ignored. They con- test. Patients may be divided into four groups.
cluded that IgG subclass interrelationships are clearly Patients with Warm IgM Autoantibodies Associa-
complex and require strictly defined populations for ted with RBC-Bound Complement. Routinely, RBCs
their study. are only tested with anti-IgG and anti-C3 (C3d). From
Dubarry and coworkers55 used a solid-phase ELAT 6% to 13% of RBCs from patients with WAIHA have
to quantitate immunoglobulin classes and IgG sub- been reported to react with anti-C3 (C3d) but not with
classes of IgG eluted from RBCs of DAT-positive anti-IgG.1,4,33 In most cases it is not known how the C3
patients. Three groups were analyzed: Group 1 came to be on the RBCs. If anti-IgM is used for the DAT,
included 23 patients with AIHA associated with warm sometimes RBC-bound IgM can be detected, and it is
autoantibodies of IgG class; group 2 included 11 assumed that the IgM autoantibody may have activated
patients without anemia but with a positive DAT; and complement causing RBC-bound C3 (C3dg). We found
group 3 included 10 healthy DAT-negative subjects. that 2% of patients with WAIHA associated with RBC-
The mean number of IgM and IgA molecules per RBC bound C3 also had RBC-bound IgM.4 Sokol and
was low in the three groups. IgG1 predominated in all coworkers58 only found 1 of 355 patients with WAIHA
groups except in two patients with AIHA, in whom to have IgM+ C3 on their RBCs. Several cases of
IgG3 made up at least 50% of total IgG. The mean WAIHA due to IgM warm autoantibodies associated
number of IgG1, IgG2, and IgG4 molecules per RBC in with a positive routine DAT due to RBC-bound C3
group 1 was about three times that in group 2, (C3d) have been reported58-67; sometimes the IgM
whereas the mean number of IgG3 molecules per RBC was only detectable by tests more sensitive than the
was 10 times as high (p < 0.001). It follows that IgG3 DAT.22-24 As the amount of anti-C3d in commercial AGS
was more common in patients of group 1, but it was is variable, these patients could present as DAT-nega-
also detected in patients of group 2. tive AIHA. IgM autoantibodies were detectable in the
Lynen and coworkers56 studied quantitation and serum of most patients and sometimes in eluates made
determination of IgG subclass of RBC-bound IgG of from the patients’ RBCs. Some investigators com-
65 patients using a newly developed gel centrifuga- mented on the difficulty of obtaining reactive eluates.
tion test (GCT) and flow cytometry (FC). The GCT Dorner and coworkers68 could only demonstrate the
involved using dilutions of the antiglobulin sera, RBC-bound IgM autoantibody if it was eluted into
including subclass antisera. The dilutions were albumin or serum. Ellis and coworkers69 studied the
selected by comparison of the GCT, flow cytometry, most suitable elution method for IgM in 42 patients
and clinical findings. The amount of IgG on the RBCs with AIHA associated with RBC-bound IgM; heat
as determined by GCT dilution cards correlated with (56°C) elution was found superior to acid stromal,
FC (r = 0.70, p < 0.0001). IgG subclass results as deter- organic solvent, or freeze-thaw methods. Although all
mined by GCT IgG subclass cards were confirmed by of the patients’ sera contained pentameric forms of IgM,
FC in 14 cases with an anti–IgG-γ chain titer ≥300, and 64% contained oligomeric forms of IgM, only pen-
whereas IgG subclass cards were not suitable in cases tameric IgM was eluted from the RBCs. Thus, it was
with anti-IgG-γ chain titers less than 300. In 44 concluded that low molecular weight IgM was not the
patients with 2+ or 3+ DAT in the GCT and anti–IgG- cause of in vivo hemolysis in these 42 patients.
γ chain titer ≤30, no hemolysis was observed, whereas Patients with Easily Detectable IgG Autoanti-
hemolysis occurred in 13 of 14 patients with an bodies, and IgM Autoantibodies that May Not Be
anti–IgG-γ chain titer ≥300. GCT data obtained by Detected by Routine Tests. If routine procedures are
IATs with anti-D sera were concordant with FC used to test the RBCs and serum of this group, the
results. They concluded that there is a correlation results associated with this group would be those most
between the amount of RBC-bound IgG and immune commonly found in idiopathic WAIHA (e.g., IgG or
hemolysis. The GCT cards that detect the anti–IgG-γ IgG+C3 detected on the RBCs; agglutination and
chain may be useful to predict hemolysis in patients perhaps lysis of enzyme-treated RBCs; a positive IAT
with a 2+ or 3+ DAT in the GCT. The diagnostic value with untreated and enzyme-treated RBCs). These
of GCT cards for IgG subclass testing needs to be results are obtained because anti-IgM is not used rou-
investigated further. tinely for the DAT and IAT, and the immunoglobulin
class of the serum antibody(ies) is (are) not usually
determined. When anti-IgM was used, we found that
WAIHA Associated with IgM RBC-bound IgM was detected on 8% of the RBCs of
Autoantibodies patients with WAIHA.4 None of the 104 patients had
only IgM on the RBCs; 2% had IgG+IgA+C3+IgM; 4%
An unusual but important catergory of WAIHA had IgG+IgM+C3; and 2% had IgM+C3. If more
consists of cases associated with IgM warm autoanti- sensitive tests are applied, then RBC-bound IgM is
bodies48,57-76 (also see Chapter 9, page 331). IgM warm detected more often. Vos and coworkers77 showed that
autoantibodies may agglutinate untreated and/or 42% of eluates prepared from the RBCs of 55 patients
enzyme-treated RBCs, hemolyze untreated and/or with WAIHA contained IgM and/or IgA in addition to
enzyme-treated RBCs, and sensitize RBCs so that IgM the IgG detected by the DAT. Using the AutoAnalyzer,
Differential Diagnosis of Immune Hemolytic Anemias 181

Hsu and coworkers19 found that 8 of 11 (73%) patients the DAT; five had a positive DAT due to RBC-bound
with WAIHA, associated with IgG-positive DATs, had complement, and two had negative DATs. Wolf and
RBC-bound IgM present in addition. None of 16 DAT- Roelcke72 also presented evidence to suggest that the
positive patients without AIHA had RBC-bound IgM. targets for IgM warm hemolysins belong to a newer
Using ELAT, Sokol and coworkers78,79 detected IgM on group of phospholipase susceptible RBC antigens and
the RBCs of 39 of 102 (38%) patients with WAIHA, that this specificity emphasizes that they are a sepa-
associated with IgG-positive DATs. rate category of warm autoantibodies.
We found that 5% of sera from patients with The relative pathogenic role of IgG versus IgM
WAIHA would agglutinate, and 0.4% would hemo- autoantibodies in patients with AIHA associated with
lyze, untreated RBCs at 37°C. Ninety percent of the the above findings is not clear, but there are some
sera agglutinated enzyme-treated RBCs, and 9% to publications73-75 where the IgM warm autoantibody
13% would hemolyze enzyme-treated RBCs, at 37°C.4 shows the characteristics associated with WAIHA asso-
We did not prove that these results were due to IgM ciated with IgM warm autoantibodies without IgG
autoantibodies, but the characteristics are what might being associated (as described in the section above),
be expected of IgM rather than IgG antibodies. von suggesting that the IgM autoantibody was pathogenic.
dem Borne and colleagues48,71 showed that autoanti- Patients with Warm IgM Autoantibodies and a
bodies that hemolyze enzyme-treated RBCs at 37°C Negative DAT. Some authors have demonstrated IgM
are IgM, and that such antibodies, by themselves, on the RBCs of patients who have AIHA but have a
cause only slight to moderate in vivo RBC destruction. negative routine DAT.18,21,70 Such cases are quite
Wolf and Roelcke61 studied eight sera containing unusual and are reviewed in Chapter 9 in the segment
warm hemolysins against enzyme-treated RBCs; none on AIHA with a negative DAT.
of these agglutinated or hemolyzed untreated RBCs at Severe (Sometimes Fatal) Hemolytic Anemia
37°C. Seven of the eight antibodies were shown to be Associated with Warm IgM Autoantibodies Direc-
IgM (one had an IgG component and one an IgA com- ted Against Determinants on Glycophorins. We
ponent in addition); one appeared to be IgG, without know of seven cases of severe WAIHA associated with
IgM or IgA. In three sera the IgM were shown to be IgM antibodies directed against determinants on
monoclonal proteins (κ light chains only). Only one of glycophorins [Ena,23 Pr,23 N,64 Ge,66 and Wrb23,74].
these eight patients had RBC-bound IgG detectable by Table 5-12 shows some data from these patients.

TABLE 5-12. RESULTS OF TESTING RBCs AND SERA FROM PATIENTS WITH WARM IgM AUTOANTIBODIES
DIRECTED AGAINST DETERMINANTS ON GLYCOPHORINS

Spontaneous
Agglutination
Case No. of Patients’ Clinical
(Ref.) DAT Eluate RBCs Serum Course

1 (70) Negative No IgM agglutinin 4+ at 37°C; (IAT ? Pure red cell aplasia.
at 37°C was negative); anti-Ena Remission of AIHA.
2 (23) 2+ (C3), no IgG IgM, 37°C, Yes IgM agglutinin 3+ at 37°C; Intravascular lysis →
(3+) agglutinin anti-EnaFS. Only reactive at hematocrit <2%; died.
low pH (e.g., pH 6.5) or in
the presence of albumin.
3 (23) 3+ (C3), no IgG, IgM, or IgA Nonreactive No IgM agglutinin 2+ at 37°C; Lymphoma. Died
37°C lysin; anti-EnaFR. Better following transfusion
reactions at low pH or in the of 5 U.
presence of albumin.
4 (23) 1/2+ C3. No IgG, IgM, or IgM, 37°C, Yes IgM agglutinin 3+ at 37°C; no Intravascular lysis.
IgA by DAT but IgM by (4+) agglutinin lysin; anti-Pr Plasma exchange.
flow cytometry. IVIG. Remission.
5 (74) 1+ (IgG + C3) Anti-Wrb reacting Not recorded IgM agglutinin 4+ at 37°C Hemoglobin dropped
by IAT (LISS); and 4+ IAT 37°C lysis from 7 g to 4 g % in
(LISS only); anti-Wrb 24 hr. Patient died.
6 (64) 4+ (C3 + IgA) (IgM by IgM, 37°C, Yes IgM agglutinin 3+ at RT (1 + at Hb 3.9 g/dL; hct 13%.
flow cytometry) anti-N 37°C). Only reactive if pH Transfused 7 units.
<7.0. Anti-N Died on day 5.
7 (66) 3+ (C3 no IgG) (IgM by IgM agglutinin- Yes No detectable antibody Hb 4.4 g/dL. Transfused
flow cytometry) ating; anti-Ge 4 units. Treated with
steroids.
Hb ↑ 13 g/dL.
Discharged on day 6.
182 Immune Hemolytic Anemias

It should be noted that the patients’ RBCs sponta- on the serology of 41 patients with AIHA associated
neously agglutinated when centrifuged in two of the with IgM warm autoantibodies. Problems were often
cases, which led to difficulties in performing DATs encountered when the patient’s RBCs were used for
and RBC typing. Treatment of the RBCs with dithio- ABO/Rh typing and/or DAT, as 61% of the RBCs
threitol (DTT) overcame the spontaneous agglutina- spontaneously agglutinated when centrifuged. This
tion but because of the effect of DTT on RBC-bound spontaneous agglutination could be overcome by
IgM, the DATs using anti-IgM were unreliable. It is treating the patient’s RBCs with DTT.82
interesting to note that following DTT treatment, with Most of the patients had a positive DAT but some
subsequent diminished spontaneous agglutination, presented as a DAT-negative AIHA.24 Most (90%) of the
IgM could sometimes still be detected (especially by patients had detectable RBC-bound C3 on their RBCs;
flow cytometry).23 This would suggest that RBC- 60% had only C3 detectable on their RBCs; 17% had
bound IgM monomers are still present following DTT IgG+ C3. Only 28% reacted with an anti-IgM standard-
treatment.23 Further work using anti-IgM by flow ized for use by the routine DAT (see Chapter 6), but
cytometry, which does not involve centrifugation of 82% of the RBCs that were nonreactive with anti-IgM
the RBCs, showed that RBC-bound IgM could be had RBC-bound IgM detected by a flow cytometric
detected by this approach.23,24,66,80 method.23,24
Several points are worth emphasizing concerning Most (91%) of the sera contained IgM, 37°C-reactive
the serology. Although these antibodies were very autoagglutins. Untreated and enzyme-treated RBCs
effective in vivo, they had unusual in vitro character- were lysed by 13% and 58% of the sera, respectively.
istics. Two of the DATs were only weakly positive, and One interesting finding was that the autoantibodies
one was negative. Although the serum antibodies were sometimes difficult to detect even though the
agglutinated RBCs strongly, they sometimes did not patient had severe (sometimes fatal) hemolytic
react by the IAT, thus they appear to be low affinity anemia. Sometimes the autoantibodies would only
antibodies susceptible to the washing process. We react or were enhanced considerably at a low pH
have also encountered this phenomenon with an IgM and/or in the presence of albumin (e.g., 30% bovine
alloanti-Ena that caused a fatal hemolytic transfusion albumin). The antibodies often reacted optimally at
reaction.81 Two of the autoanti-Ena, that caused fatal 30°C (rather than the expected 37°C). More (39%)
hemolysis, reacted only at low pH (e.g., 6.5) or in the autoantibodies had clearly defined specificities than
presence of 30% albumin.23 Although one example of are usually seen with WAIHA (see Chapter 7). Eight of
auto anti-Wrb caused fatal hemolysis, it was detected the autoantibodies had clear-cut Rh specificity (e.g.,
poorly in systems using normal ionic strength saline.74 an anti-c agglutinin), two had senescent cell antigen
Brain and coworkers76 published an interesting specificity, and there was one each showing anti-Ena,
hypothesis as to why autoantibodies directed at epi- -Wrb, -Pr, -N, -Ge, and -IT specificity.24
topes on glycophorin A were associated with such
severe hemolytic anemia. The inexplicable severity of a Characterization of Antibodies in the
WAIHA associated with anti-Pr led them to test the Cold Agglutinin Syndrome
hypothesis that the hemolysis was primarily due to a
change in the function of glycophorin A, on which the Cold agglutinins that satisfy, the “classic” characteris-
Pr antigen is located. The lectins Maclura pomifera and tics described for antibodies in CAS, often produce
wheat germ agglutinin that bind to glycophorin A striking in vitro findings and make their presence
induced hemolysis of normal RBCs in vitro. Lectin known in several sections of the clinical laboratory.
binding led to an increase in RBC membrane perme- These cold agglutinins have a titer of 1000 to 16,000 or
ability to sodium and potassium, the former resulting higher in saline at 4°C (normal is <64), and cause in
in an influx of water and subsequent hemolysis. The vitro agglutination of anticoagulated blood at room
response was glycophorin A specific as concanavalin A, temperature that is grossly evident to the naked eye.
which binds to band 3, did not cause hemolysis and Indeed, all the RBCs may be bound in one huge agglu-
peanut agglutinin only did so after removal of RBC tinate, which may be mistaken by the uninitiated as a
sialic acid. The lectin-induced cation leak was not clot (see Fig. 5-1). Preparation of adequate blood
mediated by activation of cation channels as the smears at room temperature becomes impossible
inhibitors, tetrodotoxin, amiloride, and 4,4’-diisothio- because of gross agglutination of RBCs, and blood
cyanate stilbene 2,2’-disulfonate, had no effect, sug- counts in automated blood cell counters produce non-
gesting that the hemolysis was due to exacerbation of sense numbers.83-86 Also, if a sample is sent to the
the inherent cation permeability of the RBC membrane. blood transfusion service for compatibility testing, the
A human IgAγ anti-Pr autoantibody and a mouse anti- cold agglutinins are immediately apparent and special
human glycophorin A antibody increased RBC perma- knowledge will be required to circumvent the prob-
bility to sodium. The role of glycophorin A in lems that they produce. For anyone who has observed
stabilizing and, upon aggregation, destabilizing the just one similar case previously, the diagnosis of CAS
phospholipids bilayer was discussed.76 comes immediately to mind.
Typical Serology of WAIHA Associated with IgM However, other patients with an elevated cold
Autoantibodies. Garratty and coworkers24 reported agglutinin titer have no evidence of hemolysis. Even
Differential Diagnosis of Immune Hemolytic Anemias 183

though these antibodies are clinically benign, they monly it is assumed that such cold agglutinins must
may react at room temperature, causing some of the in be potent enough to cause in vivo shortening of RBC
vitro problems previously described. The converse is survival and, if the patient has a hemolytic anemia, a
also true; that is, we frequently encounter patients diagnosis of CAS is made without further character-
who do not have striking increases in their cold ization of the antibody. However, such patients may
agglutinin titer yet appear to have CAS since they have WAIHA accompanied by a clinically insigni-
have an acquired hemolytic anemia, a DAT that is ficant cold agglutinin. Thermal amplitude studies
positive only with anticomplement sera, a moderate and other serologic tests are necessary to determine
elevation of cold agglutinin titer and thermal ampli- the correct diagnosis.
tude, and no other definable cause of hemolysis after If a patient has a positive DAT due to only comple-
extensive evaluation and long-term follow-up. ment sensitization, and very high titer cold agglutinin
Further complicating the situation is the fact that (>1000 at 4°C), CAS is very likely to be the correct
about 30% of the patients with WAIHA also have a diagnosis and, in these cases, thermal amplitude
modest to moderate elevation of cold agglutinin titer studies are not as critical. However, many cold agglu-
and/or thermal amplitude and, especially if charac- tinins in patients with CAS have a titer that is 1000 or
teristic or “typical” serologic findings of WAIHA are less at 4°C in saline (see Tables 5-13 and 5-14), and it is
not present, the patient may be erroneously diag- in these patients that determination of thermal ampli-
nosed as having CAS. tude is extremely important. Characterization of the
Therefore, the mere observance of cold autoaggluti- patient’s cold agglutinin is crucial in order to avoid
nation is not diagnostic of CAS, and when they are diagnostic errors.
encountered, the task is to determine whether the The specificity of the antibody in 54 patients with
patient has clinically insignificant albeit abnormal CAS in our series is indicated in Table 5-13. Although
cold agglutinins, has WAIHA with an associated but such specificity results are of interest, they are not
probably insignificant elevation of cold agglutinin essential either for diagnosis or for the selection of
titer or thermal amplitude, or has CAS. A rather blood for transfusion. Other specificities are discussed
common diagnostic error is overdiagnosis of CAS in in Chapter 7.
patients who have benign cold antibodies and patho- Development of Criteria to Distinguish Benign
genic warm autoantibodies. Also, in rare patients, Cold Agglutinins from Those Associated with In
characteristic findings of both WAIHA and CAS occur Vivo Hemolysis. To develop guidelines that may be of
simultaneously (see Chapter 3, “Combined Cold and value in the diagnosis of CAS, we studied various char-
Warm AIHA” and later in this chapter, page 191). acteristics of cold agglutinins in patients with CAS and
Table 5-2 shows the AIHA screen results associated in patients without hemolytic anemia. As with most
with CAS. RBC antibodies, it is difficult to predict the in vivo
Additional Comments Regarding Serologic Tests. significance from in vitro tests, and it is obvious that the
Cold agglutinins that are reactive at room tem- titer of an antibody when tested at 4°C is not a direct
perature are frequently encountered and are noticed measurement of its clinical importance.
because they cause difficulty in compatibility testing Our study characterized cold agglutinins by various
and cause agglutination visible to the naked eye in techniques, but it emphasized the reactivity of the anti-
an anticoagulated blood sample, as well as other of body at various temperatures against red cells sus-
the in vitro phenomena previously described. Com- pended in saline or in 30% albumin medium.87 The use

TABLE 5-13. COLD AGGLUTININ TITERS AND SPECIFICITIES OF 135


ANTIBODIES FROM PATIENTS WITH CAS

No. of Patients (%)

Titers San Francisco Los Angeles United Kingdom Total

<1000 (range = 8–512) 23 (43) 29 (45) 0 52 (39)


1000–8000 27 (50) 30 (46) 7 (44) 64 (47)
16,000–64,000 3 (5) 6 (9) 7 (44) 16 (12)
125,000–512,000 1 (2) 0 2 (12) 3 (2)
Total 54 65 16 135
Specificities
Anti-l 49 (91)
Anti-i 4 (7)
Unidentified 1 (2)

San Francisco: data extracted from ref. 4


United Kingdom: data extracted from ref. 1
Los Angeles: From Garratty G, unpublished data of samples submitted, 1981–2002.
184 Immune Hemolytic Anemias

TABLE 5-14. TITERS AND THERMAL AMPLITUDE (WITH OR WITHOUT ALBUMIN) IN 32 PATIENTS—
RELATIONSHIP TO HEMOLYTIC ANEMIA

4°C 30°C 37°C


Hemolytic
Patient Anemia Saline Albumin Saline Albumin Saline Albumin

1 Yes 2560 5120 1 640 0 320


2 Yes 5120 10240 1 640 0 160
3 Yes 2560 5120 4 320 0 80
4 Yes 2560 5120 20 320 0 20
5 Yes 1280 1280 10 160 1 1
6 Yes 10240 10240 1 40 0 1
7 Yes 640 1280 1 20 0 1
8 Yes 640 5120 10 160 0 10
9 Yes 640 2560 1 40 0 1
10 Yes 2560 5120 1 10 0 1
11 Yes 1280 1280 1 10 0 1
12 Yes 8192 4096 128 128 8 16
13 Yes 1024 256 0 1 0 0
14 Yes 1024 2048 0 2 0 1
15 Yes 640 2560 0 10 0 1
16 Yes 640 1280 0 40 0 1
17 Yes 1280 5120 0 20 0 1
18 Yes 1024 256 0 4 0 2
19 Yes 640 640 0 1 0 0
20 Yes 640 640 0 1 0 0
21 Yes 2048 1024 0 8 0 1
22 Yes 320 320 0 10 0 0
23 Yes 320 320 0 1 0 0
24 Yes 256 1024 1 4 0 1
25 Yes 256 512 1 2 0 0
26 Yes 256 512 0 4 0 0
27 Yes 128 1024 1 1 0 0
28 Yes 8 128 0 1 0 1
29 No 320 320 0 0 0 0
30 No 320 640 0 0 0 0
31 No 1280 2560 0 0 0 0
32 No 320 20 0 0 0 0

Data from Garratty G, Petz LD, Hoops JK: The correlation of cold agglutinin titrations in saline and albumin with haemolytic anaemia. Br J Haemat
1977;35:587–595.

of 30% albumin medium was based on the report of 30°C, and 37°C in the 32 patients who were divided
Haynes and Chaplin,88 who described an enhancing into 4 groups. Patients 1 to 12 all had hemolytic anemia
effect of albumin on cold agglutinins that was associated with high titer cold agglutinins at 4°C, and
particularly striking in one patient. This patient had a all sera reacted at 30°C with saline-suspended RBCs.
hemolytic anemia associated with only a modestly ele- The sera of two patients (5 and 12) reacted at 37°C with
vated cold agglutinin titer of 128 at 4°C using saline- saline-suspended red cells, and all 12 reacted at 37°C in
suspended RBCs, but a titer of 131,000 in the presence the presence of albumin.
of bovine albumin. We studied 32 patients who had ele- Patients 13 to 21 all had hemolytic anemia associ-
vated cold agglutinin titers and positive DATs due to ated with cold agglutinins of high titer at 4°C, but
complement sensitization of their RBCs. Twenty-eight none of the sera from these 9 patients reacted at 30°C
of these patients had an associated hemolytic anemia. unless albumin was present in the incubation mixture.
We found that testing for cold hemolysins in vitro In the presence of albumin all nine reacted at 30°C,
did not differentiate clinically important antibodies and five of the nine reacted also at 37°C.
from benign cold agglutinins. Although sera from all Patients 22 to 28 all had hemolytic anemia but
patients with CAS caused lysis of enzyme-treated normal to moderately increased cold agglutinin titers
RBCs at 20°C, sera from 3 of the 4 patients without at 4°C (i.e., not more than about two tubes above our
hemolytic anemia also caused in vitro lysis. upper limit of normal). Three of the 7 sera reacted at
The cold agglutinin titer at 4°C, when performed in 30°C with saline-suspended RBCs, and all 7 reacted in
saline or albumin, did not correlate with the presence the presence of albumin. Two of the 7 (14 and 28)
or absence of hemolysis, but determination of the reacted at 37°C in the presence of albumin.
thermal range did prove helpful.87 Table 5-14 shows the Patient 28 is of considerable interest because he had
comparative titrations using saline and albumin at 4°C, severe hemolytic anemia associated with a cold agglu-
Differential Diagnosis of Immune Hemolytic Anemias 185

tinin titer of only 8 at 4°C when saline-suspended at 4°C) and a wide thermal range (4°C to 37°C). The
RBCs were tested (normal cold agglutinin titers at 4°C patient was closely followed over a 3-year period and
range up to 64 in our laboratory). In the presence of no evidence of hemolysis was ever documented,
albumin the cold autoantibody reacted to a titer of 128 despite a persistence of a positive DAT and the cold
at 4°C and up to 37°C. These tests were reproducible autoagglutinin. Similarly, Arndt and coworkers91
even with very strict control of temperature. This reported a patient who did not have evidence of
patient had a strongly positive DAT due to comple- hemolysis although he had an auto-anti-B reacting to
ment sensitization. In addition to the cold agglutinin, a titer of 8192 at 4°C and 16 at 30°C. The anti-B caused
the serum contained a cold hemolysin, reacting opti- weak hemolysis of group B enzyme-treated but not
mally at 20°C against enzyme-treated RBCs. The DL untreated RBCs in vitro.
test was negative. The reactions at 30°C and 37°C in Sokol and coworkers92 studied 221 patients who had
the presence of albumin showed anti-I specificity, and cold autoagglutinins that reacted to ≥30°C, in the pres-
the reactions were inhibited by 2-mercaptoethanol. ence of albumin. They found they could divide them
No other antibody activity at 37°C could be demon- into three groups. Group 1 contained 116 (52%) indivi-
strated in the serum or in an eluate prepared from the duals in whom hemolysins were never detected, and
patient’s red cells. Thus, it appeared that all reactions the 74 (34%) patients in group 2 had monophasic
were due to the IgM cold autoantibody with a high hemolysins alone, whereas both monophasic and
thermal maximum and that no warm autoantibodies biphasic hemolysins were detected in the 31 (14%)
optimally reactive at 37°C were present. No other group 3 patients. There was a significantly higher pro-
cause of hemolysis was found in this patient. portion of patients in groups 2 and 3 with haptoglobin
Patients 29 to 32 all had increased cold agglutinin levels less than 0.1 g/L compared with groups 1 and 2,
titers at 4°C but no associated hemolytic anemia. respectively. DAT results showed that the autoimmune
None of these sera reacted at 30°C or 37°C, even in response became more complex and IgM predominant
the presence of albumin. Thus, all patients with through groups 1 to 3, resulting in an increasing ability
hemolytic anemia had cold agglutinins that reacted at to activate complement, which was reflected in increas-
30°C in albumin in contrast to negative reactions in ing hemolysin activity and number of patients with
patients without hemolytic anemia. Based on the active hemolysis. The 31 patients in group 3 were
empiric findings in this report, it appears that reac- mostly elderly (median age, 71 years at presentation)
tivity of a cold agglutinin at 30°C in albumin may be and the majority had chronic CAS, several in associa-
the optimal means for distinguishing clinically tion with lymphoid neoplasms or carcinomas; only four
insignificant cold agglutinins from those associated had acute CAS. It would appear that these results are in
with in vivo hemolysis. conflict with our results and conclusions,87 but it should
An additional point of significance in this study was be pointed out that the two studies were very different.
that we excluded IgG antibody activity in 22 of the 28 We studied the titers and thermal amplitude of cold
sera that reacted at 30°C in the presence of albumin by autoagglutinins in patients who were suspected of
treating them with 2-mercaptoethanol. It was con- having CAS. Sokol and coworkers92 studied random
cluded that IgG antibodies were not causing the orig- cold antibodies from patients referred to their blood
inal agglutination reactions observed at 30°C or 37°C. center for antibody investigations, from 1984 to 1996.
Isbister and coworkers89 reported three interesting The predictive value of our findings was high in our
patients who had lymphoproliferative disease with patients; we would not expect the predictive value to be
immunoglobulin M lambda (IgMλ) monoclonal pro- high in Sokol’s patient population. For instance, many
teins and severe AIHA. These three patients had cold warm autoantibodies (i.e., optimally reactive at 37°C),
autoagglutinins of low titer (8, 8, and 64) at 4°C; all three or alloantibodies will react at 30°C in the presence of
autoagglultinins reacted strongly at 30°C and weakly at albumin. Nevertheless, the findings of Sokol and
37°C. All patients had only complement detectable on coworkers92 are of interest as they showed that the
their RBCs. In two patients, splenectomy led to a remis- demonstration of hemolysins correlated better with the
sion; the third patient responded to steroids. presence of hemolytic anemia (based on lowered hapto-
It should be noted that we proposed this means of globin levels) than thermal amplitude and cold agglu-
distinguishing benign cold agglutinins from those tinin titers. As Table 5-2 shows, we found that only 2%
causing CAS more than 25 years ago. We had every or 24.5% of sera from CAS patients hemolyzed
expectation that exceptions to the “rule of thumb” that untreated or enzyme-treated RBCs, respectively, at 20°C
we suggested would be found, and that more precise without added complement. When complement was
criteria would subsequently be reported. In contrast, added (at optimal pH), then 14% and 94% of the sera
our suggested criteria have served as remarkably hemolyzed untreated or enzyme-treated RBCs, respec-
good guidelines for diagnosis. tively. So, it appears that a hemolysin test at 20°C, when
However, two exceptional patients have been performed optimally (i.e., added complement, optimal
reported who had high titer, high thermal amplitude pH, enzyme-treated RBCs), has a very good predictive
cold agglutinins but not hemolytic anemia. Sniecinski value but this is not as easy for a nonspecialist labora-
and coworkers90 reported a patient who had a cold tory to perform as it is to test for agglutination at 30°C
antibody that had both a high titer (greater than 4096 in the presence of albumin.
186 Immune Hemolytic Anemias

Essential Diagnostic Tests for CAS. On the basis of hemolytic anemia, (2) a positive DAT caused by sensi-
the preceding findings we offer the following approach tization with C3, (3) a negative DAT using anti-IgG,
to the diagnosis of CAS. The diagnosis must be consid- (4) the presence of a cold autoagglutinin with reactiv-
ered in all patients with acquired hemolytic anemia ity up to at least 30°C in saline or albumin. Although
who have a positive DAT using anti-C3 and a negative a cold agglutinin will be present to a titer of ≥256 at
DAT using anti-IgG. A practical initial serum screening 4°C, except in very unusual cases, we would empha-
procedure is to test the ability of the patient’s serum to size that the cold agglutinin titer is not the critical
agglutinate saline-suspended normal red cells at 20°C aspect of diagnosis. Indeed, a screening test can be
(or room temperature) after incubation for 30 to 60 performed in which one observes for agglutination at
minutes (Chapter 6). This recommendation is based on 30°C using a single tube with the patient’s undiluted
data illustrated in Table 5-2 namely, that the sera of serum and normal RBCs.
essentially all patients in our series who had CAS An alternative diagnosis must be sought for
caused agglutination of saline-suspended red cells at patients who do not satisfy all these criteria.
20°C. If this screening test is negative, CAS is extremely An example of typical findings in a patient with
unlikely; if positive, further studies are necessary to CAS are given in the following case report with sero-
determine the thermal amplitude of the antibody. logic findings listed in Tables 5-15A and B.
When CAS appears to be a possible diagnosis on
the basis of the preceding evaluation, studies of the P A T I E N T 1 : A 52-year-old woman sought medical
thermal range of reactivity of the antibody in saline attention because she noted that her fingers turned blue on
and albumin are indicated. It is also convenient to exposure to cold. This sign was associated with mild dis-
determine possible Ii blood group specificity of the comfort under her fingernails and a sensation of tingling.
antibody simultaneously (see Chapter 6). The titer of The findings resolved quickly upon warming. She had no
the cold agglutinin in CAS is invariably highest at 4°C other complaints and, in particular, had no symptoms of
and progressively decreases at higher temperatures. fatigue. Approximately 1 year previously her hematocrit
Of particular note are the reactions at 30°C and 37°C. was 35%. There was no history of blood loss. Site
If the former is positive in saline or albumin, the anti- appeared neither acutely nor chronically ill, and a physi-
body may well be of pathogenetic significance, that is, cal examination revealed no abnormal findings.
it may be causing short red blood cell survival in vivo. Laboratory data revealed a hemoglobin level of
If the reaction at 37°C is also positive in the presence 9.6 mg/dL, hematocrit 25%, white cells 7700/μL with a
of albumin (as was true in 19 of the 28 [68%] patients normal differential, platelets 392,000/μL, and retictulo-
in Table 5-14) or when albumin is not present (2 of 28 cytes 15.6%. Total bilirubin was 1.2 mg/dL; direct reacting
patients [7.1%]), the antibody will cause problems in bilirubin was 0.21 mg/dL, serum iron was 107, and total
the compatibility testing (see Chapter 10). iron binding capacity was 199 μg/dL. Urinalysis revealed
Using clinical information, the results of the DAT, specific gravity of 1.009, and there was no proteinuria or
and the preceding screening tests, a reasonably glycosuria; a test for Bence Jones protein was negative.
confident assessment of the presence or absence of Tests for occult blood in the stool were negative. Antinuclear
CAS may be made. CAS may be diagnosed if the fol- antibody was negative. Total protein was 7.6 g/dL; serum
lowing are present: (1) clinical evidence of acquired electrophoresis indicated that there was no M component.

TABLE 5-15A. SEROLOGIC FINDINGS IN A TYPICAL PATIENT WITH COLD AGGLUTININ SYNDROME

DAT: Polyspecific antiglobulin serum: 4+ Anti-IgG: negative Anti-C3: 3+

Screening Tests for Serum Antibody

Serum AS + AC* Control AC

20°C, Untreated Cells


Lysis 0 2+ 0
Agglutination 4+ 4+ 0
20°C, Papainized Cells
Lysis 0 4+ 0
Agglutination 4+ 4+ 0
37°C, Untreated Cells
Lysis 0 0 0
Agglutination 0 0 0
37°C, Papainized Cells
IAT 0 0 0
Agglutination 0 0 0

* AS, acidified serum; AC, acidified complement


Differential Diagnosis of Immune Hemolytic Anemias 187

TABLE 5-15B. TITER, THERMAL RANGE, AND SPECIFICITY OF COLD AGGLUTININ USING SALINE-SUSPENDED RBCs

Dilution Patient
Serum 1 2 4 8 16 32 64 128 256 512 1024 2048 4096 Titer

4°C
adult Ol 4+ 4+ 4+ 4+ 4+ 4+ 4+ 4+ 21⁄2+ 1+ 1+ 1+ 0 2048
cord Oi 4+ 4+ 4+ 4+ 4+ 4+ 31⁄2+ 2+ 2+ 1+ 1+ 0 0 1024
adult Oi 4+ 4+ 4+ 4+ 31⁄2+ 31⁄2+ 21⁄2+ 2+ 1+ 0 0 0 0 512
25°C
adult Ol 4+ 4+ 3+ 21⁄2+ 2+ 11⁄2+ 1+ (1)+ 0 0 0 0 0 128
cord Oi 3+ 1+ 0 0 0 0 0 0 0 0 0 0 0 2
adult Oi 2+ 0 0 0 0 0 0 0 0 0 0 0 0 1
30°C
adult Ol 1+ 0 0 0 0 0 0 0 0 0 0 0 0 1
cord Oi 0 0 0 0 0 0 0 0 0 0 0 0 0 0
adult Oi 0 0 0 0 0 0 0 0 0 0 0 0 0 0
37°C
adult OI 0 0 0 0 0 0 0 0 0 0 0 0 0 0
cord Oi 0 0 0 0 0 0 0 0 0 0 0 0 0 0
adult Oi 0 0 0 0 0 0 0 0 0 0 0 0 0 0

Specificity:
Serum: anti-I
Eluate: not tested
Cold agglutinin titer: 2048 vs. adult Ol RBCs at 4°C
Cold agglutinin thermal amplitude: up to 30°C
Summary: Findings characteristic of cold agglutinin syndrome
Follow-up: Chronic moderately severe anemia with no change in serologic reactions in seven years

The laboratory technologists noted that 4+ agglutina- Therapy with chlorambucil was initiated at a time when
tion occurred at room temperature (23°C) and that there her hematocrit had dropped to 18%. A bone marrow
was no agglutination at 37°C. Hemolysis was present in biopsy at this time revealed multiple nodules of mature
serum samples collected at room temperature. The DAT lymphocytes; the biopsy was interpreted as indicating the
was positive. Smears of aspirated bone marrow revealed presence of it lymphoma. However, no abnormal physical
gross clumping and were inadequate for interpretation. findings were present and, in particular, there was no
Sections revealed normoblastic erythropoiesis and no evi- hepatosplenomegaly or lymphadenopathy. Her hemat-
dence of an infiltrative process, although an occasional ocrit improved to 24% during therapy with 8 mg chlor-
focus of mature lymphocytes was seen. ambucil daily, during which time she was also more
Her serologic findings are indicated in Tables 5-15A meticulous about avoiding cold. Chlorambucil was gra-
and B. Although the antibody was only minimally reactive dually increased to 10 mg per day and she subsequently
at 30°C without albumin, it caused 4+ agglutination at that developed pancytopenia and reticulocytopenia. Her
temperature in the presence of 30% albumin and reacted hematocrit reached a nadir of 12.5%; white cells
against adult OI RBCs even at 37°C. The clinical and sero- 2500/μL; platelets 88,000/μL; and reticulocytes 2.0%.
logic findings were considered diagnostic of CAS. She was treated with transfusions of 2 units of RBCs on
Her course initially was quite stable, although she had three occasions in order to keep her hematocrit at about
exacerbations in cold weather, particularly when she also 20%, after which her marrow function spontaneously
had an upper respiratory infection. Six years later she devel- improved. Her pancytopenia resolved and her hematocrit
oped a sore throat, malaise, chilly sensations, and a fever remained stable at about 24%. A repeat bone marrow
of 101°F. Her hematocrit dropped to 16% and reticulocytes examination revealed marked improvement, and nodules
dropped to 3.5%. Site was admitted to the hospital and of lymphocytes were no longer apparent, although the total
placed in a warmed room, and she wore gloves and warm number of lymphocytes was increased above normal.
stockings continuously. A bone marrow aspiration revealed Nine years after she first presented, her disease was
striking erythroid hyperplasia with normal megakaryocytes stable and she had received no further transfusions. Her
and granulocytic precursors. Occasional plasma cells were hematocrit level remained about 24% to 26%, she
present, but there was no lymphocytic infiltration. Her hema- received no specific therapy, and she continued to work
tocrit reached a low of 14%, but then gradually improved. daily. She still noticed acrocyanosis on exposure to mild
She was discharged from the hospital when the hematocrit cold. There were no new physical findings.
reached 20%, and a progressive increase to 31% was Serologic evaluations performed in intervals throughout
noted as she was followed as an outpatient. her course revealed a minimal diminution in the strength of
In subsequent years her usual hematocrit level became the DAT using anti-C3 but the cold agglutinin titer and
somewhat lower, usually in the range of about 24%. thermal amplitude were essentially unchanged.
188 Immune Hemolytic Anemias

As we have already indicated, the ability of cold tinins. Results showed that the heavy chains of four
antibodies to cause lysis at 20°C (of enzyme-treated IgM anti-I cold agglutinins were exclusively VHI
RBCs with added complement at optimal pH) was a subgroups and their light chains were exclusively VκII
characteristic of essentially all antibodies from subgroups. In contrast, the light chains of two cold
patients with CAS, but it was also a characteristic of agglutinins with anti-Pr specificity were not VκII,
antibodies from some patients with no evidence of in whereas their heavy chains were not restricted to a
vivo hemolysis. Thus, positive tests for lysis at 20°C single subgroup. The amino acid sequences at the first
are not diagnostic of CAS. What is more important is hypervariable region of light chains (positions 25 to 35)
that cold antibodies will cause a decreasing amount of were similar in two of the four anti-I cold agglutinins.
lysis at progressively higher temperatures. If results These sequences were different from that of the light
for lysis are more strongly positive at 37°C than at chain of another cold agglutinin with anti-Pr specificity.
20°C or 30°C, a warm hemolysin that may be a sepa- These results supported the concept that only antibod-
rate antibody must be suspected. ies with the same specificity can share similar primary
structure at their antigen-combining sites.
IgG VH genes are classified into families according to
Immunochemistry and Molecular sequence homology. At first it was thought that most
Analysis of Cold Autoagglutinins of them could be assigned to three major groups: VHI,
Associated with CAS VHII, and VHIII. DNA analysis has revealed the pre-
sence of three additional VH families: VHIV, VHV, and
With rare exceptions, pathogenic cold agglutinins are VHVI. Pascual and coworkers104 and Silverman and
IgM. They exhibit a reversible, thermal-dependent Carson105 showed that anti-I cold autoagglutinins
equilibrium reaction with RBCs, with association exclusively use heavy chains that derive from the VHIV
being favored at lower temperatures. The thermal family. Pascual and coworkers104 and Silberstein and
amplitude depends on the concentration and equili- coworkers106 confirmed that pathologenic anti-I cold
brium constant of the antibody. The cold agglutinins autoagglutinins are encoded by the VH4.21 heavy
associated with chronic idiopathic CAS found in older chain gene. Pascual and coworkers104 showed that a
individuals are almost always monoclonal IgM pro- nucleotide change in H chain CDR1 results in the sub-
teins. Antibodies of the most common specificity stitution of an aspartic acid residue for glycine at posi-
(anti-I) almost always have only κ light chains; rare tion 31, suggesting that this amino acid might be
examples of anti-i often have only λ light chains.93-98 critical to the recognition of I antigen on the RBC.
The demonstration of the exclusive, or virtually exclu- Silberstein and coworkers106 studied anti-I and anti-i
sive, occurrence of κ chains in anti-I cold agglutinins cold agglutinins from B-cell clones and from the
was the first example of a relationship between anti- peripheral circulation of patients with lymphoprolifer-
body specificity and light chain type. Amino acid ative syndromes. Sequence analyses of expressed vari-
sequence studies, and the studies of the N-terminal able region genes indicated that both anti-i and anti-I
amino acid, of light chains isolated from cold agglu- specificities from B-cell clones from two patients were
tinin light chains identified glutamic acid as the encoded by the VH4.21 or a very closely related VH4
N-terminal amino acid, indicating that they belong to heavy chain gene, whereas the expressed light chain
the subgroup VκII.99,100 A restriction within the heavy genes differed. The anti-i–secreting B cells expressed
μ-chains has also been demonstrated by peptide unmutated germline-encoded VH4.21 and VκI gene
mapping heavy chains of 14 isolated cold agglu- sequences. The VH region gene encoding anti-I has the
tinins.98 Thirteen of the 14 belonged to one μ sub- closest homology (97%) to the VH 4.21 germline gene
group, as shown by the lack of a particular peptide in and differs at the protein level by only three amino
40% of monoclonal M-globulins without known anti- acids. In contrast, although VL region gene encoding
body activity.98 anti-I is most homologous (96%) to the VκIII, kv328
Further immunochemical studies of the heavy chains germline gene, there are seven amino acid differences
of IgM cold agglutinins indicated that the μ chains of due to nonrandom replacement mutations, which sug-
different cold agglutinins share antigenic determinants gests a role for antigen-mediated selection in the anti-I
not found on IgM molecules lacking cold agglutinin response of this individual. These studies were
activity. These specific antigenic determinants appea- extended by a structural survey of 20 additional cold
red to be present on the heavy chains of monoclonal agglutinins using antipeptide antibodies specific for
proteins of both anti-I and anti-i specificities,101 and determinants VH and VL regions. All anti-I and anti-i
there is one report of similar structural characteristics of cold agglutinins were shown to express VH 4 heavy
a restricted polyclonal protein from a patient with chains, and 14 of 17 cold agglutinins expressed a pre-
Mycoplasma pneumoniae infection.102 Furthermore, anti- viously described VH 4 second hypervariable region
sera specific for the heavy chains of IgM cold agglu- determinant, termed VH-4-HV2a. It was also found
tinins cross-react with heavy chains of IgA cold that 13 of 14 anti-I cold agglutinins used VκIII light
agglutinins, which suggests that there is a common chains, whereas the anti-i cold agglutinins used light
idiopathic specificity.101 chains from at least three VL families. Taken together,
Gergely and coworkers103 studied the variable the data show that anti-i and anti-I cold agglutinins
regions of four anti-I and two anti-Pr cold autoagglu- probably both derive from the VH4.21 gene (or a
Differential Diagnosis of Immune Hemolytic Anemias 189

closely related gene). Furthermore, the restricted VH as well as on neoplastic B cells secreting cold agglu-
and different VL gene use in anti-i and anti-I may tinins. The 9G4 idioptype was present on all 48
reflect the close structural relationship of the i and pathogenic anti-I/i cold agglutinins studied.113,116
I antigens. Silverman and coworkers105 demonstrated However, natural/benign cold agglutinins do not
that many anti-I have κIII L chains that express express the 9G4 idiotope.107
primary sequence-dependent Id determinants linked Thus, available data suggest a diverse B-cell origin of
to the germline gene Humkv325 or a nearly identical natural cold agglutinins, which infers that B-cell clones
gene. secreting natural cold agglutinins do not necessarily
The study of the structures of cold agglutinating represent precursors of the monoclonal B-cell expan-
autoantibodies has centered around determining gene sions secreting pathogenic cold agglutinins. The differ-
segment usage encoding the variable regions by ences between benign and pathogenic cold agglutinins
sequence analysis, and detecting the presence of vari- have not been completely characterized, and further
able region idiotopes (Id) using anti-idiotopic anti- study should provide insights into our understanding
bodies (anti-Id).107,108 It has been determined that of what makes an autoantibody pathogenic.108
there are both naturally occurring, nonpathogenic Occasionally, cold agglutinins are cryoprecip-
anti-I/i cold agglutinins, and pathogenic anti-I/i cold itable.117-125 Cryoprecipitable anti-I has been observed,
agglutinins derived from B cell clonal expansions. and at least one third of anti-i cold agglutinins are cry-
Despite attempts to correlate antibody subclass, com- oprecipitable.124 In 50 patients with CAS, the total con-
plement-fixing ability, or antibody titer with the pres- centration of IgM varied from 0.7 to 24.5 mg/mL.125 In
ence or absence of hemolysis,29,50,109,110 it remains most of the patients, the cold agglutinin accounts for
unclear what structural properties distinguish the entire elevation of serum IgM above normal
“benign” autoantibodies from hemolytic ones.107 levels. In two patients studied by Harboe,125 a homo-
Initial studies of the structure of these antibodies geneous protein remained in the serum after adsorp-
focused on the constant regions of the immunoglobu- tion with RBC stroma in the cold. This finding was
lin molecules. Later studies focused on studying the shown to be due to the presence of an additional
structural diversity of the variable regions of these monoclonal protein without cold agglutinin activity,
autoantibodies through the use of anti-idiotypic thus indicating that these were instances of biclonal
reagents and by direct nucleotide sequencing of the gammopathy.
rearranged immunoglubulin variable region genes.107 Many investigators have noted similarities of labo-
Naturally occurring IgM cold agglutinins are com- ratory findings in patients with CAS and patients
monly found in low titers and have no pathological who have monoclonal IgM proteins without cold
significance. These nonpathogenic autoantibodies are agglutinin activity. In both instances, bone marrow
thought to be the product of either random rearrange- findings may consist of increased numbers of abnor-
ment of the immunoglobulin gene segments in the mal lymphoid and plasma cells,126 although Dacie1
bone marrow and/or produced as a result of molecu- and Firkin and coworkers127 reported no excess of
lar mimicry with structures on the surface of infec- plasma cells in patients with CAS. Schubothe128 ana-
tious agents.108 These autoantibodies have been lyzed 14 patients with CAS and found 9 had bone
shown to be polyreactive and encoded by the VH3 marrow lymphocyte counts exceeding 25%; it
gene segments V3-15 and V3-48111 and VH4 family exceeded 40% in six patients. In one patient, there
gene segments including V4-49, V4-29, and V4- was an impressive progression of lymphocytic
34.111,112 Some anti-i antibodies from the normal B cell infiltration of the marrow 4 years after the onset of
repertoire were V4-34 encoded, and selected antibod- the disease. On the basis of these similarities, some
ies had similar avidities to pathogenic V4-34 encoded investigators think that a typical case of CAS is prob-
autoantibodies. Therefore, while autoantibodies with ably a variant of Waldenström’s macroglobulinemia
anti-I/i specificity are encoded by some members of in which the IgM M-component has cold agglutinin
the VH4 and VH3 families, they are not associated with activity. However, there are many differences
disease.108 between CAS and patients with macroglobulinemia
In contrast, cold agglutinins produced in chronic without cold agglutinin activity (see Chapter 3).
lymphoproliferative conditions are exclusively In 1982, Crisp and Pruzanski129 reported on patients
encoded by the V4-34 gene segment in association with cold autoagglutinins. Among 78 patients with
with different D and JH gene segments.105,113-116 The persistent cold agglutinins, 31 had lymphoma, 13 had
V4-34 encoded regions of pathogenic cold agglutinins Waldenström’s macroglobulinemia, 6 had chronic
have been found in both germline configuration and lymphocytic leukemia, and 28 had chronic CAS. The
somatically mutated. This use of a single gene average age was over 60 years. Patients with chronic
segment in antibodies directed against an auto- CAS had more hemolytic crises, bleeding, and
antigen is very rare in immunology, and suggests that Raynaud’s phenomena and less frequently lym-
the V4-34 gene segment is required to encode the anti- phadenopathy or hepatosplenomegaly. The frequency
I/i specificity.110,115 Also, a monoclonal anti-idiotypic of anemia, positive DAT test results, cryoglobulinemia,
antibody termed “9G4” has been described, which and Bence Jones proteinuria was similar in the various
recognizes an idiotypic determinant present on the groups. Survival time from diagnosis was on average
heavy chain of both anti-I and anti-i cold agglutinins 2 years in lymphoma, 2.5 years in Waldenström’s
190 Immune Hemolytic Anemias

macroglobulinemia, more than 6 years in chronic lym- gest that IgG cold antibodies are not infrequently
phocytic leukemia, and more than 5 years in chronic present in sera containing high-titer cold antibodies
CAS. Anti-I was common in chronic CAS (74%) and alongside the characteristic and usually dominant
uncommon in other groups (32% to 33%). Anti-i and IgM antibodies. Less commonly, IgA cold antibodies
other cold agglutinins were rare in chronic CAS and are present as well.
common in lymphoma and Waldenström’s macroglob- Freedman and Newlands 136 investigated two
ulinemia. In chronic cold agglutinin disease and in women, aged 23 and 30, respectively. The first gave a
Waldenström’s macroglobulinemia, cold agglutinins 10-year history of chronic AIHA and the second gave a
usually had κ light chains, 92% and 71%, respectively, 1-year history of AIHA; in both instances the onset of
whereas in lymphoma, 71% of cold agglutinins had λ hemolysis had been acute. Both cases had IgM auto-
light chains. The type of light chains related to the antibodies that were cold agglutinins with anti-I
specificity of cold agglutinins: 58% of IgM/κ were anti- specificity and were complement binding. Both cases
I and 75% of IgM/λ had other specificities. Cold agglu- also had IgG nonagglutinating autoantibodies that were
tinins were cytotoxic to autologous and allogeneic of wide thermal range and also had anti-I specificity, but
lymphocytes. Occasionally, more autologous than allo- were not complement binding. The RBCs of both
geneic cells were killed, implying that the former may patients were coated with C4/C3d and IgG
be precoated in vivo with the antibodies. Crisp and Dellagi and coworkers131 performed immunologic
Pruzanski129 concluded that conditions with persistent studies in a case of chronic AIHA associated with a
cold agglutinins are a spectrum that varies from cold agglutinin that had a titer of only 16. Despite the
“benign” chronic cold agglutinin disease to malignant low titer cold agglutinin, there was prominent autoag-
lymphoma. Marked differences in the light chain type glutination of RBCs soon after blood collection. The
of cold agglutinins, specificity toward membranous authors demonstrated that the cold agglutinin was
antigens, and severity of clinical manifestations were directed against the Pr RBC antigen and was not
noted in benign and malignant varities. reduced by mercaptoethanol treatment of the serum;
IgG and IgA Cold Autoagglutinins. Although CAS the antibody reacted up to 37°C. They concluded that
is almost always caused by an IgM autoantibody, the chronic AIHA was related to low titer monoclonal
there are a number of reports of cases in which the IgG cold agglutinins with anti-Pr activity.
cold autoagglutinin was of the IgG immunoglobulin Silberstein and coworkers137 reported a patient with
class.130-141 Included among such cases are those with IgG κ and IgM κ immunoglobulins, both possessing
biclonal antibodies, for example, a combination of IgG cold agglutinin activity. In view of the predominance
and IgM autoantibodies. Occasionally, IgA autoagglu- of the IgG cold agglutinin, splenectomy was recom-
tinins are found as well. mended for treatment and in the subsequent
Roelcke and coworkers133 described two children 30 months no additional therapy was necessary.
with transient CAS who had IgG cold agglutinins. Szymanski and coworkers138 described a 65-year-old
Mygind and Ahrons134 reported two patients with first man with severe hemolytic anemia of 2 months’ dura-
trimester abortion and idiopathic CAS. Using sucrose tion who had IgG and IgM lambda agglutinins in his
gradient ultracentrifugation and 2 mercaptoethanol serum. Their thermal optimum was unusual, the titers
treatment, the authors demonstrated the presence of IgM with adult group O cells being 16 at 4°C, 128 at 22°C,
as well as IgG cold agglutinins. Moore and Chaplin135 and 1 at 37°C. They did not bind complement in vitro,
described a patient with severe AIHA whose RBCs were consistent with the finding that the patient had negative
strongly coated with IgG and C3d. In addition to a DAT using anti-C3d. Their specificity could not be deter-
typical IgM anti-I cold agglutinin of modest titer, the mined. The patient responded to corticosteroid therapy
serum contained a lambda IgG antibody that bound and remained well without treatment 14 months after
more strongly to RBCs at 4°C (titer 64) than at 37°C (titer the hemolytic episode. The authors suggested that the
4). The patient improved following splenectomy, but the presence of IgG cold agglutinins may be predictive of a
authors were not successful in demonstrating IgG or favorable response to corticosteroid therapy.
IgM antibodies in a concentrated splenic extract. Silberstein and coworkers132 reported six patients
Ratkin and coworkers130 estimated by radial im- whose serum cold agglutinin was not inactivated or was
munodiffusion the IgG, IgA, and IgM concentration in incompletely inactivated with the IgM-reducing agent
eluates that had been prepared in a carefully stan- dithiothreitol. In five of these patients, isolation of the
dardized and controlled way from normal RBCs that antibodies revealed that two patients had predomi-
had been sensitized at 0°C by exposure to 19 different nantly IgG cold-reactive antibody, which was associated
high-titer cold antibodies, 14 of which had been with smaller amounts of IgM in one patient and with
derived from patients with CAS. As expected, the IgM IgA in the other; two patients had predominantly IgM
concentrations were 32 to 4800 times the concentra- cold agglutinin with lesser amounts of cold-reactive
tions of residual Ig predicted to be present in the fluid IgG; and one patient had an IgG cold agglutinin only.
in which the RBCs were suspended after the last Curtis and coworkers139 reported a 21-year-old man
washing. Seventeen of the 19 eluates contained 9 to with fulminant CAS who was hospitalized with
660 times the predicted concentration of IgG and 6 of hemoglobinemia, hemoglobinuria, a hemoglobin
the 19 eluates contained 12 to 226 times the expected concentration of 3.3 gm/dL, a negative DAT with poly-
concentrations of IgA. These findings certainly sug- specific and anti-C3d reagents, a negative DL test, and a
Differential Diagnosis of Immune Hemolytic Anemias 191

cold agglutinin titer of 80. He failed to respond to serology of classic WAIHA and CAS).4,39,149-155 The
corticosteroids, multiple plasma exchanges, and cyclo- second group usually have IgG and/or C3 on their
phosphamide; he required 54 transfusions in 10 days RBCs, and their sera contain IgG 37°C-reactive anti-
to maintain a hemoglobin concentration of 6.0 to bodies together with cold autoagglutinins of normal
10.0 g/dL. He improved dramatically after a splenec- titer, but in contrast to the one third of WAIHA men-
tomy was performed. The wide thermal amplitude cold tioned above, the cold autoagglutinins react at 37°C
agglutinin proved to be an IgG1 κ antibody with Pra and/or 30°C. There are only three published reports
specificity. The patient’s serum exhibited normal com- that relate to this group and the report by Sokol and
plement activation. When the DAT was carried out at coworkers155 does not give any cold agglutinin titers,
0°C to 4°C, the result was strongly positive for IgG; the so some of the patients may belong in group 1.
IAT at 0°C to 4°C was positive with the patient’s serum Shulman and coworkers154 believe that up to 8% of
diluted 1 in 640. Within 6 months, he was in complete WAIHA may belong to this group. Table 5-16 shows
remission and receiving no therapy. The patient is the serological results of 39 patients we believe fit in
remarkable for his requirement for many transfusions this group. In these patients we believe the cold
and for DATs that were consistently negative for C3d. autoagglutinin, although of normal titer, could have
Angevine and coworkers142 described a patient with played a role in the hemolytic process. It is of interest
reticulum sarcoma who had acrocyanosis associated to note that Isbister and coworkers89 described four
with cold exposure. He had a negative DAT and no patients with lymphoproliferative disease, with IgM λ
hemolytic anemia. He had a cold agglutinin with monoclonal proteins and severe AIHA, who had low-
titers of 1024 to 4096. The antibody was found to be titer high thermal amplitude cold autoagglutinins but
IgA (kappa light chains only). We described a similar no IgG autoantibodies. These findings support the
case.143 Our patient first developed symptoms of acro- findings we have that such antibodies can play a
cyanosis associated with the cold, 21 years before pathogenic role and that this role may be significant.
being diagnosed as having multiple myeloma associ-
ated with a monoclonal IgA paraprotein. He had a
negative DAT and no hemolytic anemia, but had a Laboratory Diagnosis of Paroxysmal
cold autoagglutinin with a titer of 16,000; the Cold Hemoglobinuria
specificity was anti-Pr. An IgA anti-Pr autoagglutinin ESSENTIAL DIAGNOSTIC TESTS
in a patient with myeloma was previously described
by Roelcke and Dorow in 1968.144 The IgA cold agglu- The diagnosis of PCH or the exclusion of that diagno-
tinins described by Angevine and coworkers142 and sis in the laboratory is usually considerably easier
Garratty and coworkers143 were studied further by than that of either WAIHA or CAS. The essential labo-
Roelcke144,145; both were confirmed as having anti-Pr1 ratory test is the Donath-Landsteiner (DL) test, which
specificity. Further examples of IgA anti-Pr have been is described in detail in Chapter 6, page 223). A posi-
described by Roelcke and coworkers.146,147 tive test is illustrated in Figure 3-4. A negative test
Tschirhart and coworkers148 reported a patient with excludes the diagnosis of PCH and a positive test is,
CAS whose serum contained a biclonal IgA and IgM with rare exceptions (described below), diagnostic of
gammopathy, with identical findings demonstrable in the disorder.
the patient’s RBCs eluate. The authors commented The autoantibody associated with PCH is termed a
that serum electrophoresis suggested that their biphasic hemolysin, that is to say, it sensitizes RBCs in the
patient had a monoclonal gammopathy, and only cold but only hemolyzes them when the RBCs reach
immunofixation electrophoresis demonstrated the 37°C. The diagnostic test is the DL test where RBCs are
presence of biclonal immunoglobulins. incubated with the patient’s serum at 0°C (e.g., melting
ice) and then moved to 37°C for a further incubation.
Patients Who Have Warm and Cold No lysis occurs following the incubation at 0°C, and no
Autoantibodies lysis occurs if the incubation is carried out only at 37°C.
The thermal amplitude of this antibody is usually less
As mentioned previously, approximately one third of than 20°C, that is to say, the antibody will give a posi-
WAIHA patients have cold agglutinins that can react tive DL test only when the initial incubation is <20°C;
quite strongly at room temperature but have normal stronger results will occur as the temperature of the
titer (at 4°C) and do not react at 30°C and 37°C. We do initial incubation is lowered. Rare patients have been
not think such antibodies are pathogenic, but some- described when the DL test is positive when the first
times patients with WAIHA have cold antibodies that incubation phase is as high as 32°C, or their DL anti-
react up to 30°C or above; such antibodies may be body would sensitize RBCs up to 37°C, as detected by
pathogenic. We reviewed the clinical and hematologi- the IAT (see below).156-158
cal findings of such patients in Chapter 3 (pages The autoantibody may sometimes agglutinate
79–81). Serologically, the patients fall into three RBCs in addition to giving a positive DL test.1 The
groups. The first group have IgG and C3 on their agglutination is usually of low titer (<64) at 4°C, and
RBCs, and their sera may contain IgG 37°C-reactive of low thermal amplitude (<20°C). The antibody is
antibodies together with high-titer high thermal IgG but is usually only detectable by the IAT if,
amplitude cold autoagglutinins (i.e., the combined following incubation of the patient’s serum and
TABLE 5-16. SEROLOGY OF 39 PATIENTS WITH WARM AND COLD AUTOANTIBODIES

192
IAT Autoagglutinin Titers Lysins
(Polyspecific
Patients DAT AGS) 20°C 37°C 4°C 30°C 37°C 20°C (UT/ET) 37° (UT/ET) Notes

1 IgG/C3 2+ 31⁄2+ 3+ 64 2 1 0/2+ 0/3+


2 IgG/C3 Micro+ 3+ 0 2 [2+] 0 0/0 0/0 Anti-Ena or –Pr
3 IgG/C3 1+ 4+ 1 32 [2+] [11⁄2+] 0/1+ 0/2+
4 IgG/C3/IgM 31⁄2+ 31⁄2+ 11⁄2 16 8 4 0/0 0/0
5 IgG/C3 Micro+ 21⁄2+ 0 16 1 1 0/0 0/0
6 IgG/C3/IgM Micro+ 3+ 0 32 1 0 0/2+ 0/3+
7 IgG/C3 0 31⁄2+ 0 64 1 0 0/1⁄2+ 0/0
8 IgG/C3 3+ 0/4+ (I/i) 0 2/64 (I/i) 0/16 (I/i) 0 NT NT Anti-i
9 IgG/C3 1+ 3+ 1 4 1 0 0/0 0/2+
10 IgG/C3 0 3+ 11⁄2 128 4 1 0/0 0/1+
11 IgG/C3 Micro+ 4+ 0 32 1 0 0/0 0/Trace
12 IgG/C3/IgM/IgA 2+ 4+ 2 32 4 2 0/1+ 0/2+
13 IgG/C3 3+ 4+ 1 16,000 [3+] [1+] NT NT
14 IgG/C3 0+ 21⁄2+ 0 8 1 1 0/0 0/2
15 IgG/C3 1+ 31⁄2+ 0 128/1024 (I/i) 0/1 (I/i) 0 0/3+ 0/0 Anti-i
16 IgG/C3 Micro+ 32 2 1 0/0 0/0 Probable anti-IT
17 IgG/C3 1+ 31⁄2+ 21⁄2 64 1 1 0/0 0/0
18 IgG/C3 Micro+ 4+ 0 64 4 0 0/0 0/0 IgG cold agglutinin
19 IgG/C3 0 3+ 0 16 0 0 NT NT Reactive at 30°C in presence
of albumin
20 IgG/C3 1+ 31⁄2+ 1 128 1 1 0/2 0/3+
21 C3 1+ 31⁄2+ 31⁄2 64 16 2 0/0 0/4+
22 C3 Micro+ 31⁄2+ 2 64 [2+] [2+] 0/4 0/4+ 20°C lysin titer = 1;
37°C lysin titer = 16
23 C3 Micro+ 4+ 1 128 2 2 0/0 0/2+ IgM warm + cold antibodies
24 C3 1⁄ +
2 31⁄2+ 1 8 1 1 0/1⁄2+ 0/3+
25 IgG/C3/IgM 1⁄ +
2 1+ 0 128 1 1 0/0 0/0
26 IgG/C3/IgM/IgA 1–2+ 3+ 2+ 4 2 1 0/2+ 0/4+
27 IgG/C3/IgM 1⁄ +
2 2+ 0 16 1 1 0/2+ 0/3+
28 IgG/C3 21⁄2+ 11⁄2+ 1+ 16–32 1 1 0/1+ 0/0
29 IgG/C3/IgM 11⁄2+ 1+ 0 32 1 0 0/0 0/0
30 IgG/C3/IgM/IgA 2+ 4+ 0 128 1 1 tr/2+ 0/1+
31 IgG/C3/IgM 1+ 3+ 1+ 32 1 1 0/0 0/0
32 IgG 3+ 1+ 0 1 [21⁄2+] [11⁄2] 0/0 0/0 Low affinity IgG anti-Pr (reactions
at 30°C only in presence of
albumin at low pH)
33 C3 0 3+ 11⁄2+ 16 4 2 0/1+ 0/4+
34 IgG/C3/IgM 0 2–31⁄2+ 1+ 16 1 1 0/0 0/trace
35 IgA 1–2+ (anti-IgA) 3+ 21⁄2+ 8 4 2 0/0 0/0
36 IgG 0 4+ 0 2 0 0 0/0 0/0 Low affinity RBC-bound IgG
antibody. Agglutination at
30°C in presence of albumin.
37 IgG/C3/IgM 1+ 21⁄2+ 11⁄2+ 32 1 1 0/1+ 0/31⁄2+
38 IgG/C3/IgM/IgA 1+ 21⁄2+ 0 32 2 NT 0/0 0/0
39 IgG/C3 0 21⁄2+ 1⁄ +
2 8 2 ? 0/0 0/0

From Garratty G, Arndt P, Leger R: Previously unpublished data.


UT = untreated RBCs; ET = enzyme-treated RBCs; [ ] = reaction with undiluted serum; titer not available.
Differential Diagnosis of Immune Hemolytic Anemias 193

RBCs at 0°C, the RBCs are washed with ice-cold During the next 24 hours he was anuric. At this point his
saline and ice-cold antiglobulin serum is used. hemoglobin was 7.0 g/dL; the hematocrit 19.6%; reticulo-
Indeed, Dacie1 has found that the IAT was a more cytes 1.1%; and the white count was 17,400 with 59% neu-
sensitive way of demonstrating antibody activity tronphils, 2% bands, 29% lymphocytes, 8% monocytes, and
than looking for lysis. Such agglutination tests must 2% eosinophils. The platelet count was 214,000/μl. The
be carefully controlled because many sera give peripheral blood film showed moderate numbers of sphe-
positive results under these circumstances if a rocytes and poikilocytes. The blood urea nitrogen was 192
polyspecific antiglobulin serum is used, due to the mg, and the creatinine 6.0 mg/dL. Total bilirubin was
presence in human sera of the normal incomplete 2.3 mg/dL, and haptoglobin 20 mg/dL. The DAT was 3+.
cold antibody. It is important, therefore, to be sure Cultures of blood, urine, throat, stool, and cerebral spinal
that monospecific anti-IgG antiglobulin serum is fluid failed to reveal pathogenic bacteria.
used. Serologic findings are summarized in Table 5-17 and
Since PCH is quite rare, one may justifiably ques- were diagnostic of PCH.
tion the advisability of performing a DL test routinely The hospital course was marked by 10 days of olig-
in patients with acquired hemolytic anemia. Our own uria necessitating peritoneal dialysis twice. Thereafter,
attitude is to be liberal with the indications for per- the urine output was normal (>60 mL/kg/day), and his
formance of the test since it is simple to perform and creatinine decreased from a high of 12.9 to 1.9 mg/dL
its inclusion avoids diagnostic errors. We certainly feel at the time of discharge. Therapy with prednisone was
that the performance of the test is indicated in any begun on the fourth hospital day at a dose of
child, any patient with hemoglobinuria, patients with 2 mg/kg/day for 1 week, after which it was gradually
a history of hemolysis exacerbated by cold, and in all decreased and then discontinued. The patient’s hemo-
cases with “atypical” serologic findings. If positive globin level dropped to 4.3 g/dL on the day after
results are obtained in the DL test, determination of admission, and he was transfused with 95 mL of P+
the specificity of the autoantibody is indicated. All RBCs without clinical symptoms of a reaction and with
cases of PCH we have encountered in the last 30 years transient benefit. Two days later his hemoglobin had
were associated with anti-P specificity. Sokol and again dropped to 4.8 g/dL, and he received 160 mL
coworkers159 found anti-P specificity in 27 of 30 (90%) of RBCs, which raised his hemoglobin to 8.6 g/dL. The
patients with PCH; specificity was not clearly defined following day his reticulocyte count was 5.1%, and
in the other three cases. Nevertheless, rare reports of thereafter his hemoglobin level remained stable and he
other specificities said to be associated with PCH have required no further transfusions.
been reported (see below and Chapter 7, page 254). Follow-up examination 2 months later revealed a hemo-
RBCs necessary for determining anti-P specificity are globin level of 13.3 g/dL, hematocrit 38.6%, reticulo-
rare but, with the assistance of reference laboratories, cytes 38.6%, creatinine 0.4 mg/dL, urinalysis normal,
specificity testing can be carried out. It should be and a 24-hour protein excretion of 50 mg.
noted that the DL test must be used to determine
specificity. Cautions Regarding the Interpretation of the DL
Typical findings in a patient with PCH are pre- Test. The DL test is essentially diagnostic for PCH, but
sented in the following case report and in Table 5-17. one must be cautious when using sera from patients
with CAS. This is true because about 15% of sera from
P A T I E N T 2 : A 41⁄2-year-old boy was in good health patients with CAS contain monophasic cold hemo-
until 10 days prior to admission to the hospital, at which lysins that will hemolyze untreated RBCs at around
time he developed a cough and sore throat. Three days 20°C. Up to 95% of such sera will cause direct lysis of
prior to admission, he developed a fever of 102°F, enzyme-treated red cells at 20°C.4 During the per-
headache, myalgia, abdominal pain, vomiting, and diar- formance of the DL test, there is a brief period of time
rhea. He was treated with oral penicillin. Two days prior to when cells and serum are at room temperature after
admission he began passing dark urine, and the next day being moved from the ice bath to a 37°C water bath.
he was noted to be jaundiced. His parents noted that his Dacie1 has reported slight hemolysis in the DL test
urine output was scanty. using 2 sera containing high-titer cold agglutinins
On admission to the hospital he was not in acute (titers of 4000 and 64,000, respectively).
distress. Temperature was 37°C, pulse 120, respirations We retrospectively tested 20 sera from patients with
30, and blood pressure 110/50 mg Hg. Physical exam- CAS who had monophasic cold hemolysins, against
ination was unremarkable. His hematocrit was 31.9%, untreated red cells, and found three (15%) of them to
bilirubin 2.4 mg/dL total and 0.2 mg/dL direct reacting, give false weakly positive DL tests. In all other aspects
and the lactic dehydrogenase (LDH) 2868 units. The these patients had symptoms and serology that was
urine was brown, specific gravity 1.017, pH 5.5, hemo- typical of CAS, rather than PCH. It should be empha-
globin test 3+, protein 3+, bilirubin negative, and only 2 sized that many other patients with CAS did not have
RBCs were present per high-power field. The creatinine positive DL tests, even when powerful monophasic
was 2.6 mg/dL, the sodium was 132 mEQ, potassium cold lysins were present in their sera. It should also be
5.9 mEQ, chloride 102 mEQ, and the carbon dioxide noted that the monophasic lysins were detected in the
8 mEQ/L. presence of fresh complement at pH 6.5 whereas the DL
194 Immune Hemolytic Anemias

unusual cases. Clinically, similarities result from the


TABLE 5-17. SEROLOGIC FINDINGS IN A PATIENT fact that acrocyanosis and hemoglobinuria may
WITH PCH develop in either disorder after exposure to cold. In
addition, both syndromes have been reported to occur
Direct Antigloublin Test: Polyspecific Antiglobulin Serum: 2+ after Mycoplasma pneumoniae infection, although PCH
Anti-IgG: negative Anti-C3: 2+
following infection with this organism is rare.
Screening Tests for Serum Antibody In the laboratory, some exceptions to the typical
findings listed in Table 5-15 occur. For example,
Serum AS + AC* Control AC Alone patients with the CAS may have a cold agglutinin titer
less than 500 (see Table 5-14). The thermal range of the
20°C, Untreated Cells
Lysis 0 0 0
autoantibody has been reported as high as 32°C to 37°C
Agglutination 0 0 0 in three exceptional cases,156-158 and there are several
reports of DL antibodies reactive at temperatures of
20°C, Papainized Cells
Lysis 0 1+ 0
18°C to 25°C.159–161 These antibodies do not require
Agglutination 1+ 1+ 0 incubation at 0°C as in the DL test and may cause
“monophasic” lysis at room temperature. There are
37°C, Untreated Cells
Lysis 0 0 0
rare reports of the specificity of the DL antibody being
Agglutination 0 0 0 reported as anti-I or anti-HI, similar to that frequently
found in CAS; we suspect that these patients may have
37°C, Papainized Cells
IAT 0 0 0
had false-positive DL tests due to monophasic lysins.
Agglutination 0 0 0 Although the preceding findings may seem to add
an air of uncertainty to the differentiation between the
*AS, acidified serum; AC, acidified complement two disorders, it is rare that the distinction is actually
Specificity of serum and eluate:
Serum: anti-P difficult, either on clinical grounds or in the labora-
Eluate: not done tory. Clinically, PCH is an acute hemolytic anemia
Cold agglutinin titer: normal
Donath-Landsteiner test: positive
associated with marked constitutional symptoms;
Summary: Findings diagnostic of paroxysmal cold hemoglobinuria modern case reports describe the disorder almost
Follow-up: Rapid improvement in anemia and in renal function after initial exclusively in children or young adults, and it is
episode of intravascular hemolysis and renal failure. Two months later the
DAT was weakly positive with anti-C3 (titer 16, score 7) and the Donath- almost always transient. In contrast, CAS generally
Landsteiner test was negative. occurs in middle-aged or elderly persons, the patient’s
symptoms are frequently just those of anemia, and the
test was performed without acidification of the serum. disorder is chronic except in the minority of patients
If enzyme-treated RBCs are used for the DL test, as sug- who have transient CAS following Mycoplasma pneu-
gested by some investigators,3 the false-positive rate moniae infection or other infectious diseases.
might be higher. We caution that if enzyme-treated red In the laboratory, a positive DL test should be con-
cells are used for the DL test, a control for monophasic sidered diagnostic of PCH unless a cold agglutinin
lysis set up in parallel should be mandatory. that is of high thermal amplitude and is a potent
Performing the DL Test in Patients with Hemoglo- hemolysin is present. When such antibodies cause
binemia. It may be impossible to determine if in vitro weakly positive reactions in the DL test, the reactions
hemolysis has occurred in the DL test if the patient’s should be interpreted as a “false-positive” DL test in a
serum is red because of marked hemoglobinemia. In patient with CAS.
this case, a simple procedure is to perform the cold Opposite findings occur in patients with PCH. That
phase of the DL test using the patient’s serum, but after is, even if the thermal range of a DL antibody is high
incubation at 4°C, carefully replace the patient’s serum
with fresh normal acidified serum before moving to the
37°C phase of the test. As a control, a similar tube can be
kept at 4°C after replacing the patient’s serum with TABLE 5-18. COMPARISON OF TYPICAL
normal serum, or one may replace the patient’s serum CHARACTERISTICS OF THE ANTIBODY IN COLD
with inactivated serum before the 37°C phase. Also, if it AGGLUTININ SYNDROME WITH THE
is known that the specificity of the antibody is anti-P DONATH-LANDSTEINER ANTIBODY OF
and P-negative RBCs are available, they may be used in PAROXYSMAL COLD HEMOGLOBINURIA
the DL test as a control. Another simple technical aid is
to compare the size of the RBC buttons following cen- Cold Agglutinin Donath-Landsteiner
trifugation. If these approaches fail, then a “cold IAT” Syndrome Antibody
can be performed. Titer (4°C) High (>500) Moderate (<64)
Comparison of PCH and CAS. CAS and PCH are Thermal range High (>30°C) Moderate (<20°C)
generally quite distinct disorders, and typical findings Bithermic lysis Negative Positive
in each syndrome are compared in Table 5-18. A few (Donath-Landsteiner test)
Immunoglobulin class IgM IgG
similar features may result in the blurring of the dis- Specificity Anti-I or i Anti-P
tinction between the two disorders, especially in some
Differential Diagnosis of Immune Hemolytic Anemias 195

enough to result in monophasic lysis, maximal lysis tested except those that were PP1Pk negative and Pk
will occur if an incubation at 0°C precedes incubation positive; thus, the specificity was anti-P. After treat-
at 37°C. Also, specificity tests are usually of value ment of the patient’s serum with 2-mercaptoethanol, it
in separating the two disorders, and the antibody in still gave a positive DL test and reacted at 37°C with
PCH is of the IgG immunoglobulin class, in contrast anti-IgG by the IAT, indicating that the autoantibody
to the antibody in CAS, which is almost always IgM. was IgG. Nordhagen158 described two similar cases in
A further means of distinguishing these disorders is which the serum gave a positive DL test with anti-P
that in PCH the peripheral blood film frequently specificity. In both patients, the antibodies also reacted
reveals striking RBC adherence and erythrophagocy- by the IAT at 37°C when the tests were read with anti-
tosis by neutrophils, whereas this we have not found IgG. Sabio and coworkers169 also reported a patient
this to be true in CAS. Further tests in the laboratory whose serum contained a biphasic hemolysin with
will reveal that RBCs from patients with PCH will be anti-P specificity, and which also reacted by IAT at
phagocytosed by monocytes in an in vitro monocyte 37°C. The patients described by the above authors all
monolayer assay (see Chapter 3, page 76). had clinical findings typical of PCH.
Lau and coworkers170 reported a patient in whom
Autoantibodies with Unusual erythrophagocytosis was noted on a routine blood
smear. The hematocrit subsequently decreased from
Characteristics in Patients Who Have 30% to 23% in 24 hours and hemoglobinemia and
Been Diagnosed as Having PCH hemoglobinuria became evident. The DL test was
The characteristic DL antibody causes hemolysis in strongly positive with both the patient’s serum and
the classic biphasic hemolysin test, is of the IgG the RBC eluate, and was inhibited by globoside but
immunoglobulin class and has anti-P specificity. not by other glycosphingolipids. The highest temper-
These laboratory findings in association with the ature of the initial erythrocyte sensitization was 10°C.
typical clinical findings described above allow for a The autoantibody agglutinated P-positive reagent
confident diagnosis of PCH. However, some antibo- RBCs incubated in LISS medium at 22°C and in the
dies in patients in whom a diagnosis of PCH has been antiglobulin phase. Further testing confirmed anti-P
made do not have anti-P specificity, and/or are IgM, specificity of the antibody. Both heat and ether
or have other unusual characteristics. eluates agglutinated P-positive but not P-negative
RBCs at 4°C and 22°C, but failed to react at 37°C and
in the antiglobulin phase.
DL Antibodies with Specificity
Other Than Anti-P IgM Antibodies Giving a Positive DL Test
Bird and coworkers160 suggested that the definition of Rarely, patients with IgM agglutinating antibodies with
PCH be restricted to cases in which the autoantibody biphasic hemolytic properties have been diagnosed as
demonstrated specificity within the P blood group having PCH. Nakamura and coworkers163 reported a
system, usually anti-P. Dacie stated1 that, in his view, patient with a biphasic hemolysin of the IgM im-
anti-P specificity is an essential criterion for labeling munoglobulin class with anti-I specificity, Ramos and
an autoantibody as a DL antibody. However, biphasic coworkers171 reported a patient with a positive DL test
antibodies showing other blood group specificities whose serum contained a single IgM cold autoantibody
have been described including anti-I,162-164 anti-p165 with IT and P specificities (anti-ITP), and Zamora and
(which may be anti-Gd), anti-“Pr-like,”166 anti-i,167 coworkers172 reported a patient with an IgM aggluti-
and anti-HI.168 The IgG nature of the anti-p,165 one nating antibody with biphasic hemolytic properties
anti-I,162 the anti-i,167 and anti-“Pr-like”166 were having anti-P specificity. Lippman and coworkers173
confirmed, thus lending support to their being also reported a patient with a biphasic anti-IgM auto-
classified as DL antibodies. hemolysin. It is difficult to determine whether or not
these cases represent instances in which an IgM cold
DL Antibodies with Unusual Activity agglutinin with hemolytic properties caused a false-
positive DL test, as described above.
Although, as mentioned above, DL antibodies com-
monly cause sensitization to agglutination by antiglo-
bulin serum when tested at 4°C with anti-IgG,1 some Antibodies with Unique Characteristics
DL antibodies have unusual properties. Lindgren Even more atypical are the following. Bastrup-
and coworkers157 described a patient whose serum Madsen and Petersen174 reported a case of benign
gave a strongly positive DL test when the initial incu- idiopathic monoclonal gammopathy with cold sensi-
bation phase was 0°C or 20°C, but was negative at tivity due to the presence of an IgG monoclonal
30°C and 37°C. The serum also reacted strongly by IAT protein that gave a positive DL test. The patient had
at 37°C with anti-IgG. The 37°C tests were repeated episodes of hemoglobinuria and developed urticarial
several times with strict control of the temperature at wheals in cold weather. On one occasion Bence Jones
all phases of testing. The serum reacted with all RBCs proteinuria was demonstrated. That the gammopathy
196 Immune Hemolytic Anemias

was of the benign type was supported by the fact that 2. Wolach B, Heddle N, Barr RD, Zipursky A, Pai KR, Blajchman
no rise was seen in the M-protein level or plasma cell MA: Transient Donath-Landsteiner haemolytic anaemia. Br J
Haematol 1981;48:425–434.
count at a follow-up examination 61⁄2 years after the 3. Heddle NM: Acute paroxysmal cold hemoglobinuria.
onset of the cold sensitivity. Transfus Med Rev 1989;3:219–229.
von dem Borne and coworkers175 reported a case of 4. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias,
AIHA associated with IgM monoclonal cold auto- 1st ed. New York: Churchill Livingstone, 1980.
5. Croucher BEE, Crookston MC, Crookston JH: Delayed hae-
agglutinins and monophasic hemolysins with anti-P molytic transfusion reactions simulating auto-immune haemo-
specificity. lytic anaemia. Vox Sang 1967;12:32.
Judd and coworkers have described an example of a 6. Croucher BEE: Differential diagnosis of delayed hemolytic
pH-dependent autoanti-P176 and two examples of transfusion reaction. In Bell CA (ed): A Seminar on Laboratory
autoanti-P that were reactive only at low ionic Management of Hemolysis. Washington, DC: American
Association of Blood Banks, 1979:151–160.
strength.177 7. Worlledge SM: The interpretation of a positive direct antiglo-
Mensinger and coworkers178 reported a patient who bulin test. Brit J Haemat 1978;39:157–162.
developed a fatal case of hemolytic anemia. Her 8. Kaplan HS, Garratty G: Predictive value of direct antiglobulin
serum contained an IgM anti-P agglutinin in combi- test results. Diagnostic Med 1985;8:29–32.
9. Garratty G: The significance of IgG on the red cell surface.
nation with an IgG anti-P monophasic lysin that Transfus Med Rev 1987;1:47–57.
reacted up to 32°C. However, the presence of a bipha- 10. Jeannet M: Specificity of the antiglobulin test in “auto-immune”
sic lysin could not be confirmed. Thus, this is a unique hemolytic anemias. Helv Med Acta 1966;33:151–163.
case in which an antibody of anti-P specificity caused 11. Gerbal A, Homberg JC, Rochant H, Perron L, Salmon C:
lysis in vitro, but did not give a positive DL test. Autoantibodies in acquired hemolytic anemia. I. Analysis of
234 cases. Nouv Rev Fr Hematol 1968;8:155–157.
Comments on the definition of PCH. By far the 12. Gerbal A, Homberg JC, Rochant H, Perron L, Salmon C: The
most common case of PCH has characteristic clinical auto-antibodies of acquired hemolytic anemias. II. Nature,
and laboratory findings. However, uncertainty arises specificity, clinical interest and mechanism of formation.
when findings are atypical, as in the cases cited above. Nouv Rev Fr Hematol 1968;8:351–368.
13. Dacie JV, Worlledge SM: Auto-immune hemolytic anemias. In
For example, some authors1,160 indicate that a diagno- Brown EB, Moore CV (eds): Progression in Hematology. New
sis of PCH is not warranted unless an antibody of York: Grune & Stratton, 1969:82–120.
anti-P specificity can be demonstrated, whereas other 14. Garratty G, Petz LD: The significance of red cell bound
investigators have reported cases as examples of PCH complement components in development of standards and
even though the biphasic hemolysin had specificity quality assurance for the anti-complement components of
antiglobulin sera. Transfusion 1976;16:297–306.
other than anti-P. 15. Petz LD, Garratty G: Complement in immunohematology.
When an IgM antibody gives a positive DL test, the Prog Clin Immunol 1974;2:175–190.
possibility that the hemolysis in vitro was actually a 16. Kerr RO, Dalmasso AP, Kaplan ME: Erythrocyte-bound C5
false-positive test caused by a monophasic hemolysin and C6 in autoimmune hemolytic anemia. J Immunol
1971;107:1209–1210.
must be carefully excluded. Even if false-positive tests 17. Salama A, Bhakdi S, Mueller-Eckhardt C, et al: Deposition of
are excluded, should patients with IgM antibodies be the terminal C5b-9 complement complex on erythrocytes by
diagnosed as having PCH, especially if the antibodies human red cell autoantibodies. Br J Haematol 1983;55:161–169.
demonstrate specificity other than anti-P and the clin- 18. Salama A, Bhakdi S, Mueller-Eckhardt C: Evidence suggesting
ical findings are not typical? the occurrence of C3-independent intravascular immune
hemolysis. Transfusion 1987;27:49–53.
Still other investigators report DL antibodies that 19. Hsu TC, Rosenfield RE, Burkart P, Wong KY, Kochwa S: Instru-
are monoclonal, antibodies that react only under very mented PVP-augmented antiglobulin tests. II. Evaluation of
special in vitro testing conditions, or antibodies with acquired hemolytic anemia. Vox Sang 1974;26:305–325.
anti-P specificity that do not cause biphasic hemolysis. 20. Garratty G, Petz LD: An evaluation of commercial anti-
globulin sera with particular reference to their anticomple-
Thus, there seems to be no strong consensus as to ment properties. Transfusion 1971;11:79–88.
what it is that defines PCH. Among the cases with 21. Garratty G: Autoimmune hemolytic anemia. In Garratty G
atypical features described above, we feel most com- (ed): Immunobiology of transfusion medicine. New York:
fortable in accepting a diagnosis of PCH when the Dekker, 1994:493–521.
antibody gives a characteristic biphasic hemolysis test 22. Arndt P, Garratty G: Use of flow cytometry for detection of
IgM on RBCs showing spontaneous agglutination (abstr).
and is of the IgG immunoglobulin class, even if the Transfusion 1994;34:68S.
specificity is not the characteristic anti-P. If the anti- 23. Garratty G, Arndt P, Domen R, et al: Severe autoimmune
body also causes agglutination, and/or reacts by IAT hemolytic anemia associated with IgM warm autoantibodies
at 20°C to 37°C, this would not negate a diagnosis of directed against determinants on or associated with glyco-
phorin A. Vox Sang 1997;72:124–130.
PCH. However, it is probably best to not definitively 24. Garratty G, Arndt P, Leger R: Serological findings in
diagnose PCH when the clinical findings are atypical autoimmune hemolytic anemia associated with IgM warm
and the antibody is IgM or has unique characteristics. autoantibodies (abstr). Blood 2001;98:61a.
25. Talano J-AM, Johnson ST, Friedman KD, et al: Serologic char-
acteristics of three cases of IgA mediated autoimmune
R E F E R E N C E S hemolytic anemia (AIHA) confirmed by flow cytometry
(abstr). Blood 2002;100:283a.
1. Dacie JV: The Haemolytic Anaemias. Part II. Auto-Immune Hae- 26. Merry AH, Thomson EE, Rawlinson VI, Stratton F: Quan-
molytic Anaemias, 2nd ed. London: J & A Churchill Ltd., 1962. titation of IgG on Erythrocytes: Correlation of number of IgG
Differential Diagnosis of Immune Hemolytic Anemias 197

molecules per cell with the strength of the direct and indirect 50. Garratty G: Factors affecting the pathogenicity of red cell
antiglobulin tests. Vox Sang 1984;47:73–81. auto- and alloantibodies. In Nance SJ (ed): Immune
27. Hazlehurst DD, Hudson G, Sokol RJ: A quantitative ELISA for Destruction of Red Blood Cells. Arlington, VA: American
measuring red cell-bound immunoglobulins. Acta Haematol Association of Blood Banks, 1989:109–169.
1996;95:112–116. 51. Dacie JV: Autoimmune hemolytic anemia. Arch Intern Med
28. Kumpel BM: A simple nonisotopic method for the quantitation 1975;135:1293–1300.
of red cell-bound immunoglobulin. Vox Sang 1990;59:34–38. 52. Engelfriet CP, von dem Borne AEG, Beckers D, et al: Immune
29. Garratty G, Nance SJ: Correlation between in vivo hemolysis destruction of red cells. In Bell CA (ed): A Seminar on
and the amount of red cell-bound IgG measured by flow Immune-Mediated Cell Destruction. Washington, DC:
cytometry. Transfusion 1990;30:617–621. American Association of Blood Banks, 1981:93–130.
30. Dubarry M, Charron C, Habibi B, Bretagne Y, Lambin P: 53. Engelfriet CP, Overbeeke MA, dem Borne AE: Autoimmune
Quantitation of immunoglobulin classes and subclasses of hemolytic anemia. Semin Hematol 1992;29:3–12.
autoantibodies bound to red cells in patients with and 54. Sokol RJ, Hewitt S, Booker DJ, et al: Erythrocyte autoanti-
without hemolysis. Transfusion 1993;33:466–471. bodies, subclasses of IgG and autoimmune haemolysis.
31. Jeje MO, Blajchman MA, Steeves K, et al: Quantitation of red Autoimmunity 1990;6:99–104.
cell-associated IgG using an immunoradiometric assay. 55. Dubarry M, Charron C, Habibi B, et al: Quantitation of
Transfusion 1984;24:473–476. immunoglobulin classes and subclasses of autoantibodies
32. Chaplin H, Nasongkla M, Monroe MC: Quantitation of red bound to red cells in patients with and without hemolysis.
blood cell-bound C3d in normal subjects and random hospi- Transfusion 1993;33:466–471.
talized patients. Br J Haematol 1981;48:69–78. 56. Lynen R, Krone O, Legler TJ, et al: A newly developed gel
33. Chaplin H Jr: Clinical usefulness of specific antiglobulin centrifugation test for quantification of RBC-bound IgG anti-
reagents in autoimmune hemolytic anemias. Progr Hematol bodies and their subclasses IgG1 and IgG3: Comparison with
1973;8:25–49. flow cytometry. Transfusion 2002;42:612–618.
34. Chaplin H, Avioli LV: Grand rounds: Autoimmune hemolytic 57. Salama A, Mueller-Eckhardt C: Autoimmune haemolytic
anemia. Arch Intern Med 1977;137:346–351. anaemia in childhood associated with noncomplement
35. Fischer JT, Petz LD, Garratty G, Cooper NR: Correlations binding IgM autoantibodies. Br J Haematol 1987;65:67–71.
between quantitative assay of red cell-bound C3, serologic 58. Sokol RJ, Booker DJ, Stamps R: Autoimmune hemolytic
reactions, and hemolytic anemia. Blood 1974;44:359–373. anemia caused by warm-reacting IgM-class antibodies.
36. Rosse WF: Quantitative immunology of immune hemolytic Immunochemistry 1998;14:53–58.
anemia. I. The fixation of C1 by autoimmune antibody and het- 59. Freedman J, Newlands M, Johnson CA: Warm IgM anti-IT
erologous anti-IgG antibody. J Clin Invest 19791;50:727–733. causing autoimmune haemolytic anaemia. Vox Sang
37. Rosse WF: Quantitative immunology of immune hemolytic 1977;32:135–142.
anemia. II. The relationship of cell-bound antibody to hemo- 60. Freedman J, Wright J, Lim FC, et al: Hemolytic warm IgM
lysis and the effect of treatment. J Clin Invest 1971;50:734. autoagglutinins in autoimmune hemolytic anemia. Transfu-
38. Worlledge SM, Blajchman MA: The autoimmune haemolytic sion 1987;27:464–467.
anaemias. Br J Haematol 1972;23(Suppl.):61. 61. Wolf MW, Roelcke D: Incomplete warm hemolysins. I. Case
39. Sokol RJ, Hewitt S, Stamps BK: Autoimmune haemolysis: An reports, serology, and immunoglobulin classes. Clin Immunol
18-year study of 865 cases referred to a regional transfusion Immunopathol 1989;51:55–67.
centre. Br Med J (Clin Res Ed) 1981;282:2023–2027. 62. Araguas C, Martin-Vega C, Massague I, de Latorre FJ:
40. Lalezari P, Louie JE, Fadlallah N: Serologic profile of “Complete” warm hemolysins producing an autoimmune
alphamethyldopa-induced hemolytic anemia: Correlation hemolytic anemia. Vox Sang 1990;59:125–126.
between cell-bound IgM and hemolysis. Blood 1982;59:61–68. 63. Hamilton T, Hoffer J, Reid ME, et al: Transient IgM autoanti-
41. Ben Izhak C, Shechter Y, Tatarsky I: Significance of multiple body with apparent anti-c specificity (abstr). Transfusion
types of antibodies on red blood cells of patients with positive 1993;33:545.
direct antiglobulin test: A study of monospecific antiglobulin 64. Garratty G, Arndt P, Tsuneta R: Fatal hemolytic anemia asso-
reactions in 85 patients. Scand J Haematol 1985;35:102–108. ciated with antoanti-N (abstr). Transfusion 1994;34:205.
42. Sokol RJ, Hewitt S, Booker DJ, et al: Enzyme linked direct 65. Friedmann AM, King KE, Shirey RS, et al: Fatal autoimmune
antiglobulin test in patients with autoimmune haemolysis. hemolytic anemia in a child due to warm-reactive immuno-
J Clin Pathol 1985;38:912–914. globulin M antibody. J Pediatr Hematol Oncol 1998;20:502–505.
43. Sokol RJ, Hewitt S, Booker DJ, Bailey A: Red cell autoanti- 66. Sererat T, Veidt D, Arndt P, et al: Warm autoimmune hemo-
bodies, multiple immunoglobulin classes, and autoimmune lytic anemia associated with an IgM autoanti-Ge. Immuno-
hemolysis. Transfusion 1990;30:714–717. chemistry 1998;14:26–29.
44. Gottsche B, Salama A, Mueller-Eckhardt C: Donath- 67. Nowak-Wegrzyn A, King KE, Shirey RS, Chen AR,
Landsteiner autoimmune hemolytic anemia in children: A McDonough C, Lederman HM: Fatal warm autoimmune
study of 22 cases. Vox Sang 1990;58:281–286. hemolytic anemia resulting from IgM autoagglutinins in an
45. Arndt PA, Leger RM, Garratty G: Serology of antibodies to infant with severe combined immunodeficiency. J Pediatr
second- and third-generation cephalosporins associated with Hematol Oncol 2001;23:250–252.
immune hemolytic anemia and/or positive direct antiglobu- 68. Dorner IM, Parker CW, Chaplin H Jr: Autoagglutination
lin tests. Transfusion 1999;39:1239–1246. developing in a patient with acute renal failure: Charac-
46. Budman DR, Steinberg AD: Hematologic aspects of systemic terization of the autoagglutinin and its relation to transfusion
lupus erythematosus: Current concepts. Ann Intern Med therapy. Br J Haematol 1968;14:383–394.
1977;86:220–229. 69. Ellis JP, Sokol RJ: Detection of IgM autoantibodies in eluates
47. Mongan ES, Leddy JP, Atwater EC, Barnett EV: Direct from red blood cells. Clin Lab Haematol 1990;12:9–15.
antiglobulin (Coombs) reactions in patients with connective 70. Brunt D, Greenfield T, Simon K, et al: An auto anti-Ena
tissue diseases. Arthritis Rheum 1967;10:502–508. mimicking an allo anti-Ena associated with pure red cell
48. von dem Borne AEG Jr, Engelfriet CP, Beckers D, et al: aplasia (abstr). Transfusion 1983;23:408.
Autoimmune haemolytic anaemias. II. Warm haemolysins– 71. von dem Borne AEG Kr, Engelfriet CP, Reynierse E, Beckers D,
serological and immunochemical investigations and 51Cr van Loghem JJ: Autoimmune haemolytic anaemia. VI. 51
studies. Clin Exp Immunol 1969;4:333–343. Chromium survival studies in patients with different kinds of
49. Nance S, Garratty G: Subclass of IgG on red cells of donors warm autoantibodies. Clin Exp Immunol 1973;13:561–571.
and patients with positive direct antiglobulin tests (abstr). 72. Wolf MW, Roelcke D: Incomplete warm hemolysins. II.
Transfusion 1983;23:413. Corresponding antigens and pathogenetic mechanisms in
198 Immune Hemolytic Anemias

autoimmune hemolytic anemias induced by incomplete warm 97. Capra JP, Kehoe JM, Williams RC Jr, et al: Light chain
hemolysins. Clin Immunol Immunopathol 1989;51:68–76. sequences of human IgM cold agglutinins. Proc Natl Acad Sci
73. Shirey RS, Kickler TS, Bell W, Little B, Smith B, Ness PM: Fatal USA 1972;69:40.
immune hemolytic anemia and hepatic failure associated with 98. Cooper AG, Chavin SI, Franklin FC: Predominance of a single
a warm-reacting IgM autoantibody. Vox Sang 1987;52:219–222. mu chain subclass in cold agglutinin heavy chains.
74. Dankbar DT, Pierce SR, Issitt PD, et al: Fatal intravascular Immunochemistry 1970;7:479.
hemolysis associated with autoanti-Wrb (abstr). Transfusion 99. Edman P, Cooper AG: Amino acid sequence at the N-terminal
1987;27:534. end of a cold agglutinin kappa chain. Fed Eur Biochem Soc
75. McCann EL, Shirey RS, Kickler TS, Ness PM: IgM autoagglu- Lett 1968;2:33.
tinins in warm autoimmune hemolytic anemia: A poor 100. Cohen S, Cooper AG: Chemical differences between indi-
prognostic feature. Acta Haematol 1992;88:120–125. vidual cold agglutinins. Immunology 1968;15:93.
76. Brain MC, Prevost JM, Pihl CE, Brown CB: Glycophorin 101. Williams RC Jr: Cold agglutinins: studies of primary structure,
A-mediated haemolysis of normal human erythrocytes: serologic activity, and antigenic uniqueness. Ann NY Acad Sci
Evidence for antigen aggregation in the pathogenesis of 1971;190:330.
immune haemolysis. Br J Haematol 2002;118:899–908. 102. Jacobson LB, Longstreth GF: Clinical and immunologic fea-
77. Vos GH, Petz LD, Fudenberg HH: Specificity and tures of transient cold agglutinin hemolytic anemia. Am J Med
immunoglobulin characteristics of autoantibodies in acquired 1973;54:514.
hemolytic anemia. J Immunol 1971;106:1172–1176. 103. Gergely J, Wang AC, Fudenberg HH: Chemical analyses
78. Sokol RJ, Hewitt S, Booker DJ, et al: Small quantities of ery- of variable regions of heavy and light chains of cold
throcyte bound immunoglobulins and autoimmune haemoly- agglutinins. Vox Sang 1973;24:432–440.
sis. J Clin Pathol 1987;40:254–257. 104. Pascual V, Victor K, Lesz D, et al: Nucleotide sequence analy-
79. Sokol RJ, Hewitt S, Booker DJ, et al: Enzyme linked direct sis of the V regions of two IgM cold agglutinins. J Immunol
antiglobulin tests in patients with autoimmune haemolysis. J 1991;146:4386.
Clin Pathol 1985;38:912–914. 105. Silverman GJ, Carson DA: Structural characterization of
80. Garratty G, Postoway N, Nance S, Arndt P: Detection of IgG human monoclonal cold agglutinins: Evidence for a distinct
on red cells of patients with suspected direct antiglobulin test primary sequence-defined VH4 idiotype. Eur J Immunol
negative autoimmune hemolytic anemia (AIHA). Book of 1990;20:351.
Abstracts of the ISBT/AABB Joint Congress, 87. 1990 (abstr). 106. Silberstein LE, Jefferies LC, Goldman J, et al: Variable region
81. Postoway N, Anstee DJ, Wortman M, Garratty G: A severe gene analysis of pathologic human autoantibodies to the
transfusion reaction associated with anti-EnaTS in a patient related i and I red blood cell antigens. Blood 1991;78:2372.
with an abnormal alpha-like red cell sialoglycoprotein. 107. Silberstein LE: B-Cell Origin of Cold Agglutinins. In Atassi
Transfusion 1988;28:77–80. MZ (ed): Immunobiology of Proteins and Peptides VII. New
82. Reid ME: Autoagglutination dispersal utilizing sulphydryl York: Plenum Press, 1994:193–205.
compounds. Transfusion 1978;18:353–355. 108. Potter KN: Molecular characterization of cold agglutinins.
83. Hattersley PG, Gerard PW, Caggiano V, Nash DR: Erroneous Transfus Sci 2000;22:113–119.
values on the Model S Coulter Counter due to high titer cold 109. Rosse WF, Adams JP: The variability of hemolysis in the cold
autoagglutinins. Am J Clin Pathol 1971;55:442–446. agglutinin syndrome. Blood 1980;56:409–416.
84. Petrucci JV, Dunne PA, Chapman CC: Spurious erythrocyte 110. Schreiber AD, Herskovitz BS, Goldwein M: Low-titer cold-
indices as measured by the model S Coulter counter due to hemagglutinin disease: Mechanism of hemolysis and
cold agglutinins. Am J Clin Pathol 1971;56:500–502. response to corticosteroids. N Engl J Med 1977;296:1490–1494.
85. DeLange JA, Eernisse GJ, Veltkamp JJ: Cold agglutinins and 111. Jefferies LC, Carchidi CM, Silberstein LE: Naturally occurring
the Coulter Counter Model S. Am J Clin Pathol 1972; anti-i/I cold agglutinins may be encoded by different VH3
58:599–600. genes as well as the VH4.21 gene segment. J Clin Invest
86. Lawrence C, Zozicky O: Spurious red-cell values with the 1993;92:2821–2833.
Coulter Counter. N Engl J Med 1983;309:925–926. 112. Schutte ME, van Es JH, Silberstein LE, Logtenberg T: VH4.21-
87. Garratty G, Petz LD, Hoops JK: The correlation of cold agglu- encoded natural autoantibodies with anti-i specificity mirror
tinin titrations in saline and albumin with haemolytic those associated with cold hemagglutinin disease. J Immunol
anaemia. Br J Haemat 1977;35:587–595. 1993;151:6569–6576.
88. Haynes CR, Chaplin H Jr: An enhancing effect of albumin on 113. Pascual V, Victor K, Randen I, et al: Nucleotide sequence
the determination of cold hemagglutinins. Vox Sang 1971;20: analysis of rheumatoid factors and polyreactive antibodies
46–54. derived from patients with rheumatoid arthritis reveals
89. Isbister JP, Cooper DA, Blake M, et al: Lymphoproliferative diverse use of VH and VL gene segments and extensive vari-
disease with IgM Lambda monoclonal protein and autoim- ability in CDR-3. Scand J Immunol 1992;36:349–362.
mune hemolytic anemia. Am J Med 1978;64:434–440. 114. Pascual V, Victor K, Spellerberg M, Hamblin TJ, Stevenson FK,
90. Sniecinski I, Margolin K, Shulman I, Oien L, Meyer E, Branch Capra JD: VH restriction among human cold agglutinins: The
DR: High-titer, high-thermal-amplitude cold autoagglutinin VH4-21 gene segment is required to encode anti-I and anti-i
not associated with hemolytic anemia. Vox Sang 1988;55:26–29. specificities. J Immunol 1992;149:2337–2344.
91. Arndt P, Do G, Garratty G, Kuriyan M, Strair R: A high titer, 115. Leoni J, Ghiso J, Goni F, Frangione B: The primary structure of
high thermal amplitude auto anti-B associated with acro- the Fab fragment of protein KAU, a monoclonal immunoglo-
cyanosis but no obvious hemolytic anemia. Transfusion 2003; bulin M cold agglutinin. J Biol Chem 1991;266:2836–2842.
43:1133–1137. 116. Grillot-Courvalin C, Brouet JC, Piller F, et al: An anti-B cell
92. Sokol RJ, Booker DJ, Stamps R, et al: Cold haemagglutinin autoantibody from Wiskott-Aldrich syndrome which recog-
disease: clinical significance of serum hemolysins. Clin Lab nizes i blood group specificity on normal human B cells. Eur J
Haematol 2000;22:337–344. Immunol 1992;22:1781–1788.
93. Harboe M, Lind K: Light chain type of transiently occurring 117. Poldre P, Pruzanski W, Chiu HM, et al: Fulminant gangrene in
cold haemagglutinins. Scand J Haematol 1966;3:269. transient cold agglutinemia associated with Escherichi coli
94. Harboe M, Furth R, van Schubothe H, et al: Exclusive occur- infection. Can Med Assoc J 1985;132:261–263.
rence of κ-chains in isolated cold agglutinins. Scand J 118. Crisp D, Pruzanski W: B-cell neoplasms with homogeneous
Haematol 1965;2:259. cold-reacting antibodies (cold agglutinins). Am J Med
95. Cooper AG, Worlledge SM: Light chains in chronic cold 1982;72:915–922.
hemagglutinin disease. Nature 1967;214:799. 119. Pruzanski W, Cowan PH, Parr DM: Clinical and immuno-
96. Feizi T: Lambda chains in cold agglutinins. Science 1967; chemical studies of IgM cold agglutinins with lambda type
156:1111. light chains. Clin Immunol Immunopathol 1974;2:234–245.
Differential Diagnosis of Immune Hemolytic Anemias 199

120. Ferriman DG, Dacie JV, Keele KD, et al: The association of 145. Roelcke D: Serological studies on the Pr1/Pra antigens using
Raynaud’s phenomena, chronic haemolytic anaemia, and the dog erythrocytes: Differentiation of Pra from Pr1 and detec-
formation of cold antibodies. Quart J Med 1951;20:275. tion of a Pr1 heterogeneity: Pr1h/Pr1d. Vox Sang 1973;24:354.
121. Rorvik K: The syndrome of high-titre cold haemagglutina- 146. Roelcke D: Specificity of IgA cold agglutinins: Anti-Pr1. Eur J
tion; a survey and a case report. Acta Med Scand 1954; Immunol 1973;3:206–212.
148:299. 147. Roelcke D, Hack H, Kreft H, et al: IgA cold agglutinins recog-
122. Gaddy CG, Powell LW: Raynaud’s syndrome associated with nize Pr and Sa antigens expressed on glycophorins.
idiopathic cryoglobulinemia and cold agglutinins: Report of a Transfusion 1993;33:472–475.
case and discussion of classification of cryoglobulinemia. 148. Tschirhart DL, Kunkel L, Shulman IA: Immune hemolytic
Arch Intern Med 1958;102:468. anemia associated with biclonal cold autoagglutinins. Vox
123. Umlas J, Kaufman M, MacQueston C, et al: A cryoglobulin Sang 1990;59:222–226.
with cold agglutinin and erythroid stem cell suppressant 149. Moake JL, Schultz DR: Hemolytic anemia associated with
properties. Transfusion 1991;31:361–364. multiple alloantibodies and low serum complement. Am J
124. Pruzanski W, Shumak KH: Biologic activity of cold-reacting Med 1975;58:431–437.
autoantibodies (first of two parts). N Engl J Med 1977; 150. Crookston JH: Hemolytic anemia with IgG and IgM autoanti-
297:538–542. bodies and alloantibodies. Arch Intern Med 1975;135:1314–1315.
125. Harboe M: Cold auto-agglutinins. Vox Sang 1971;20:289–305. 151. Freedman J, Lim FC, Musclow E, et al: Autoimmune
126. Ritzman SE, Levin WC: Cold agglutinin disease: A type of haemolytic anaemia with concurrence of warm and cold red
primary macroglobulinemia: A new concept. Tex Rep Biol cell autoantibodies and a warm hemolysin. Transfusion
Med 1962;20:236. 1985;25:368–372.
127. Firkin BG, Blackwell JB, Johnston GA: Essential cryoglobulin- 152. Kajii E, Miura Y, Ikemoto S: Characterization of autoantibo-
aemia and acquired haemolytic anaemia due to cold agglu- dies in mixed-typed autoimmune hemolytic anemia. Vox
tinins. Aust Ann Med 1959;8:151. Sang 1991;60:45–52.
128. Schubothe H: The cold hemagglutinin disease. Semin 153. Patten E, Reuter FP: Evan’s syndrome: possible benefit from
Hematol 1966;3:27. plasma exchange. Transfusion 1980;20:589–593.
129. Crisp D, Pruzanski W: B-cell neoplasms with homogeneous 154. Shulman IA, Branch DR, Nelson JM, et al: Autoimmune
cold-reacting antibodies (cold agglutinins). Am J Med hemolytic anemia with both cold and warm autoantibodies.
1982;72:915–922. JAMA 1985;253:1746–1748.
130. Ratkin GA, Osterland CK, Chaplin H Jr: IgG, IgA, and IgM 155. Sokol RJ, Hewitt S, Stamps BK: Autoimmune haemolysis:
cold-reactive immunoglobulins in 19 patients with elevated mixed warm and cold antibody type. Acta Haematol 1983;
cold agglutinins. J Lab Clin Med 1973;82:67–78. 69:266–274.
131. Dellagi K, Brouet JC, Schenmetzler C, Praloran V: Chronic 156. Ries CA, Garratty G, Petz LD, et al: Paroxysmal cold hemoglo-
hemolytic anemia due to a monoclonal IgG cold agglutinin binuria: Report of a case with an exceptionally high thermal
with anti-Pr specificity. Blood 1981;57:189–191. range Donath-Landsteiner antibody. Blood 1971;38:491–499.
132. Silberstein LE, Berkman EM, Schreiber AD: Cold hemagglu- 157. Lindgren S, Zimmerman S, Gibbs F, Garratty G: An unusual
tinin disease associated with IgG cold-reactive antibody. Ann Donath-Landsteiner antibody detectable at 37°C by the
Intern Med 1987;106:238–242. antiglobulin test. Transfusion 1985;25:142–144.
133. Roelcke D, Anstee DJ, Jungfer H, Nutzenadel W, Webb AJ: 158. Nordhagen R: Two cases of paroxysmal cold hemoglobinuria
IgG-type cold agglutinins in children and corresponding anti- with a Donath-Landsteiner antibody reactive by the indirect
gens: Detection of a new Pr antigen: Pra. Vox Sang 1971;20: antiglobulin test using anti-IgG. Transfusion 1991;31:190–191.
218–229. 159. Sokol RJ, Booker DJ, Stamps R: Erythropoiesis: paroxysmal
134. Mygind K, Ahrons S: IgG cold agglutinins and first trimester cold haemoglobinuria: A clinico-pathological study of
abortion. Vox Sang 1972;23:552–560. patients with a positive Donath-Landsteiner test. Hematology
135. Moore JA, Chaplin H Jr: Autoimmune hemolytic anemia 1999;4:137–164.
associated with an IgG cold incomplete antibody. Vox Sang 160. Bird GW, Wingham J, Martin AJ, et al: Idiopathic nonsyphilitic
1973;24:236–245. paroxysmal cold haemoglobinuria in children. J Clin Pathol
136. Freedman J, Newlands M: Autoimmune haemolytic anaemia 1976;29:215–218.
with the unusual combination of both IgM and IgG autoanti- 161. Vogel JM, Hellman M, Moloshok RE: Paroxysmal cold hemo-
bodies. Vox Sang 1977;32:61–68. globinuria of nonsyphilitic etiology in two children. J Pediatr
137. Silberstein LE, Shoenfeld Y, Schwartz RS, Berkman EM: A 1972;81:974–977.
combination of IgG and IgM autoantibodies in chronic cold 162. Bell CA, Zwicker H, Rosenbaum DL: Paroxysmal cold hemo-
agglutinin disease: Immunologic studies and response to globinuria (P.C.H.) following mycoplasma infection: Anti-I
splenectomy. Vox Sang 1985;48:105–109. specificity of the biphasic hemolysin. Transfusion 1973;13:
138. Szymanski IO, Teno R, Rybak ME: Hemolytic anemia due to 138–141.
a mixture of low-titer IgG lambda and IgM lambda agglu- 163. Nakamura H, Watanabe T, Hayashida T, Ichimaru M: Donath-
tinins reacting optimally at 22°C. Vox Sang 1986;51: Landsteiner antibody of the IgM class with anti-I specificity
112–116. and possible efficacy of azathioprine therapy in paroxysmal
139. Curtis BR, Lamon J, Roelcke D, Chaplin H: Life-threatening, cold hemoglobinuria: A case report. Rinsho Ketsueki
antiglobulin test-negative, acute autoimmune hemolytic 1990;31:1548–1552.
anemia due to a noncomplement-activating IgG1 kappa cold 164. Green ES, Dvenish A, Bradshaw HH, Davis SG, England JM,
antibody with Pra specificity. Transfusion 1990;30:838–843. Contreras M: Chronic haemolysis caused by an unusual
140. Goldberg LS, Barnett EV: Mixed gamma G-gamma M cold Donath-Landsteiner-like (DL) antibody associated with
agglutinin. J Immunol 1967;99:803–809. non-Hodgkin’s lymphoma (NHL). Transfusion Med 1990;1
141. Ambrus M, Bajtai G: A case of an IgG-type cold agglutinin (Suppl. 1):15.
disease. Haematologia 1969;3:225–235. 165. Engelfriet CP, Beckers D, von dem Borne AE, et al:
142. Angevine CD, Anderson BR, Barnett EV: A cold agglutinin of Haemolysins probably recognizing the antigen p. Vox Sang
the IgA class. J Immunol 1966;96:578. 1972;23:176–181.
143. Garratty G, Petz LD, Brodsky I, et al: An IgA high-titer cold 166. Judd WJ, Wilkinson SL, Issitt PD, et al: Donath-Landsteiner
agglutinin with an unusual blood group specificity within the hemolytic anemia due to an anti-Pr-like biphasic hemolysin.
Pr complex. Vox Sang 1973;25:32–38. Transfusion 1986;26:423–425.
144. Roelcke D, Dorow W: Besonderheiten der Reaktionswerte 167. Shirey RS, Park K, Ness PM, et al: An anti-i biphasic hemo-
eines mit Plasmocytom-gA-Paraprotein identischen lysin in chronic paroxysmal cold hemoglobinuria. Transfusion
Kälteagglutinins. Klin Wschr 1968;46:126. 1986;26:62–64.
200 Immune Hemolytic Anemias

168. Bell CA, Zwicker H: Donath-Landsteiner hemolysin with 174. Bastrup-Madsen P, Petersen SH: Monoclonal gammapathy
anti-HI specificity. Transfusion 1967;7:384. with the M-component behaving like Donath-Landsteiner
169. Sabio H, Jones D, McKie VC: Biphasic hemolysin hemolytic haemolysin. Scand J Haematol 1971;8:81–85.
anemia: Reappraisal of an acute immune hemolytic anemia of 175. von dem Borne AE, Mol JJ, Joustra-Maas N, et al: Auto-
infancy and childhood. Am J Hematol 1992;39:220–222. immune haemolytic anaemia with monoclonal IgM (kappa)
170. Lau P, Sererat S, Moore V, et al: Paroxysmal cold hemoglobin- anti-P cold autohaemolysins. Br J Haematol 1982;50:345–350.
uria in a patient with Klebsiella pneumonia. Vox Sang 1983; 176. Judd WJ: A pH-dependent auto-agglutinin with anti-P
44:167–172. specificity. Transfusion 1975;15:373–376.
171. Ramos RR, Curtis BR, Eby CS, et al: Fatal autoimmune 177. Judd WJ, Steiner EA, Capps RD: Autoagglutinins with appar-
hemolytic anemia (AHA) associated with IgM bi-thermic anti- ent anti-P specificity reactive only by low-ionic-strength salt
P and cold IT. Joint Congress of the International Society of techniques. Transfusion 1982;22:185–188.
Blood Transfusion and the American Association of Blood 178. Mensinger E, Lerner W, Leger R, et al: Serological profile asso-
Banks Book of Abstracts. S341. 1990. ciated with a fatal case of paroxysmal cold hemoglobinuria
172. Zamora C, Hernandez-Jorda M, Herrero S, et al: Donath- (abstr). Transfusion 1995;35:21S.
Landsteiner antibody detectable at 37°C by the antiglobulin
test in a HIV+ patient. Transfusion Med 1998;8:278.
173. Lippman SM, Winn L, Grumet FC, et al: Evans’ syndrome as
a presenting manifestation of atypical paroxysmal cold hemo-
globinuria. Am J Med 1987;82:1065–1072.
C H A P T E R 6

The Serologic
Investigation of
Autoimmune Hemolytic
Anemia

In this chapter, we review the AIHA is made by determining the in vitro characteris-
laboratory tests that are of tics of the autoantibody. Often, the differential diag-
value in the diagnosis of auto- nosis can be achieved in the laboratory by simple
immune hemolytic anemia extensions of tests that are used every day in the blood
(AIHA). The rationale for per- bank. The majority of cases of AIHA can be correctly
formance of the tests and the diagnosed simply by taking into account the clinical
interpretation of the results findings, the direct antiglobulin test (DAT) using
are discussed briefly, and the anti-IgG and anti-C3 (containing anti-C3d,g or C3d), a
methods are described in cold agglutinin titer, and a simple antibody screening
detail. Each of these tests has a procedure using careful technique (e.g., strict control
place in the diagnostic evaluation of at least some of incubation temperatures). However, such abbrevi-
patients with acquired hemolytic anemia, but some of ated evaluations lead to a surprising number of mis-
the procedures described in this chapter are indicated interpretations and erroneous diagnoses. Therefore,
only infrequently. Information in this chapter serves as when we encounter a case of hemolytic anemia sus-
an extension of the discussion in Chapter 5 of the dif- pected of having an immune basis, we prefer, as a
ferential diagnosis of the AIHAs. In Chapter 5, no tech- routine, to perform a panel of screening tests and then
nical details are given, but instead a more detailed to perform more specific tests to develop a more
narrative description of the step-by-step approach to confident diagnosis.
diagnosis is presented. Additional technical procedures Red cell autoantibodies, like alloantibodies, are
that are relevant to the investigation of drug-induced usually IgM or IgG; on occasion they may be IgA. In
immune hemolytic anemia and compatibility testing vitro, the IgM antibodies will usually directly aggluti-
are described in Chapters 8 and 10, respectively. nate saline-suspended red blood cells (RBCs) (e.g., the
The AIHAs can be classified into three main groups: IgM anti-I associated with cold agglutinin syndrome
(1) those associated with warm autoantibodies (i.e., [CAS]); often they will hemolyze RBCs if the condi-
optimally reactive at 37°C); (2) those associated with tions are right. For instance, one may have to use
cold agglutinins (optimally reactive at 0°C to 4°C, but enzyme-treated RBCs and/or add fresh complement
with a wide thermal range); and (3) those associated to demonstrate the lysis. IgG autoantibodies do not
with a biphasic cold hemolysin, the Donath- often cause direct agglutination of saline-suspended
Landsteiner antibody (i.e., paroxysmal cold hemoglo- normal RBCs but almost always agglutinate enzyme-
binuria [PCH]). The diagnosis of the specific type of treated RBCs or agglutinate untreated RBCs when
201
202 Immune Hemolytic Anemias

potentiators (e.g., polyethylene glycol) are present. human globulin. More recently, monoclonal AGS that
IgG autoantibodies rarely cause in vitro lysis of RBCs does not contain heterophil antibodies is being used.
(the IgG auto anti-P biphasic hemolysin, associated Thus, if the human globulin, in the form of antibody,
with PCH, is an exception) but often sensitize RBCs is attached to the RBC membrane, the AHG will com-
and can be detected by the antiglobulin test (AGT). bine with it, crosslinking the sensitized RBCs and
causing agglutination (Fig. 6-1).
As shown in Figure 6-1, anti-IgG reacts mainly with
THE ANTIGLOBULIN TEST the Fc portion (i.e., heavy chains) of human IgG mol-
ecules. Antibodies to light chains may be present in
In 1945, Coombs and coworkers1 demonstrated the polyclonal AGS as well; in a polyspecific AHG, this
presence of nonagglutinating Rh alloantibodies (the has no disadvantages and theoretically may be an
“incomplete” or “sensitizing” antibodies) in sera, by advantage by forming extra “bridges” across adjacent
the antiglobulin reaction. light chains. In a monospecific anti-IgG, the presence
of antibodies to light chains can lead to false-positive
results because light chains are shared by IgG, IgM,
Principles of the Antiglobulin Test and IgA; only the heavy chains are specific for each
Red cell autoantibodies and alloantibodies are gamma class.
globulins, usually IgM or IgG. If they are IgM, they In 1946, Boorman and coworkers2 and Loutit and
usually directly agglutinate saline-suspended RBCs. Mollison3 reported that the RBCs from a number of
In contrast, IgG antibodies often do not agglutinate patients with idiopathic acquired hemolytic anemia
RBCs but react with the corresponding antigens on reacted with AHG. The first application of the test (i.e.,
the RBC membrane, giving a “sensitized” RBC. Thus, detection of Rh antibodies in serum)1 became known as
chemically speaking, the RBCs are sensitized with the indirect antiglobulin test (IAT), and the second
gamma globulin. application (i.e., detection of in vivo sensitization)2,3
Coombs and coworkers1 postulated that if a rabbit became known as the DAT. Table 6-1 shows where
was injected with human gamma globulin, the rabbit these two tests can be applied in immunohematology.
would form antibodies to the foreign protein, that is, In 1947, Coombs and Mourant4 showed that the
anti-human globulin (AHG). This antiglobulin serum component in AGS that reacted with Rh-sensitized
(AGS) after suitable processing (e.g., adsorption of RBCs could be inhibited from reacting by the addition
heterophil antibodies) would react specifically with of small amounts of human gamma globulin; alpha
and beta globulins did not inhibit the reaction. Dacie5
reported that RBCs from various patients with AIHA
reacted differently in the AGT. Some patients reacted
better with high dilutions of the AHG (i.e., exhibited a
Antihuman lgG
prozone effect), whereas others reacted better with
more concentrated AHG. In addition, the addition of
purified gamma globulin to the AHG did not inhibit
the reaction of the AHG with the RBCs from some
patients. The reactions that could be inhibited
appeared to be those associated with warm autoanti-
bodies. In contrast, those not inhibited appeared to be
Human lgG associated with high titer cold autoagglutinins; it was
red cell antibody

TABLE 6-1. USES OF THE ANTIGLOBULIN TEST

A. Direct antiglobulin test; valuable in diagnosis of


1. Autoimmune hemolytic anemia
2. Drug-induced immune hemolytic anemia
3. Hemolytic transfusion reactions due to alloantibodies
4. Hemolytic disease of the fetus and newborn
B. Indirect antiglobulin test; useful for
1. Detection of antibodies in serum
(a) Antibody detection
Red cell (b) Crossmatching
2. Detection of antibodies in eluates prepared from sensitized
FIGURE 6-1. The antiglobulin test. Antihuman IgG in the antiglobulin RBCs
serum reacts with red cell bound IgG antibody, which causes the 3. Determination of specificity of antibodies
development of a lattice formation and agglutination. Although the 4. Typing of RBCs
reaction is illustrated with IgG-sensitized cells, similar reactions can (a) Testing for weak D phenotypes
occur with red cells coated with other proteins (e.g., complement (b) Using antiglobulin reactive typing reagents (e.g.,
components), provided that appropriate antibodies are present in the anti-Fya, anti-Jka, etc.)
antihuman serum.
The Serologic Investigation of Autoimmune Hemolytic Anemia 203

suggested that the RBCs from such patients were factured for immunologic use can be used after appro-
probably sensitized with antibodies that were priate standardization (see later).
non–gamma globulins. Further experiments6 showed
that the non–gamma globulin sensitizing the RBCs Significance of a Positive DAT
was not antibody but complement. The role of com-
plement in immunohematology, and the AGT in par- The results of a DAT should reflect what is happening
ticular, has been reviewed previously.7-10 in vivo, not what has occurred in vitro through the use
Some immune hemolytic anemias are caused by of inappropriate techniques. For instance, the RBCs
immunoglobulins other than IgG. However, IgM sen- from refrigerated clots obtained from normal indi-
sitization of RBCs is difficult to detect with the viduals often have a positive DAT; this sometimes also
AGT11,12; furthermore, IgM antibodies that cause occurs when testing RBCs from patients’ clotted
immune hemolytic anemia characteristically, if not samples left at room temperature. This is commonly
invariably, fix complement, which is much more due to the presence of a naturally occurring cold non-
readily detected.13 IgA antibodies only infrequently agglutinating autoantibody (“normal incomplete cold”
play a role in RBC sensitization, and in such cases, antibody), which has anti-H specificity and binds com-
other immune globulin and/or complement compo- plement to the RBCs.13,19 The commonly found low titer
nents are almost always, although not invariably, anti-I cold autoagglutinin also plays an additional role
found on the RBC surface as well.14-16 Thus, it does in autocomplement sensitization at 4°C. Because of this
not seem reasonable to demand standards for manu- phenomenon, the results of a positive DAT on RBCs
facturers that would require the presence of anti-IgM from a cooled clotted sample, when using an anti-
and anti-IgA as a component of polyspecific AGS, complement-containing reagent, should never be
although we prefer to have them present. Interested accepted without confirming the results on either
workers, especially those in research and reference freshly drawn blood kept at 37°C or in EDTA anti-
laboratories, will continue to be concerned about the coagulant; the EDTA prevents (by binding calcium
unusual instances in which they are of use, but such needed for C1 activation) any in vitro complement
needs can usually be met by the use of monospecific binding but does not interfere with any in vivo bound
antisera (carefully standardized for use with RBCs) in complement.
selected cases. A positive DAT, even when due to in vivo sensitiza-
To summarize, polyspecific AHG used for the tion, does not necessarily indicate AIHA, or even that
investigation of AIHA must contain anti-IgG and an autoantibody has caused the positive DAT.20,21
anti-C3d (or anti-C3d,g) and may contain antibodies Positive DATs can be obtained (even when warm
to other C3 determinants (e.g., C3b, C3c) and C4; anti- [37°C] blood or EDTA blood is used) because of many
bodies to other immunoglobulins (e.g., IgA or IgM) causes; some are listed here.
would be a bonus.
Causes of Positive DAT
Monospecific Antiglobulin Reagents 1. Autoantibodies against intrinsic RBC antigens,
Although it is reasonable and perhaps most conven- leading to sensitization of the RBCs with immunoglob-
ient to perform the initial DAT with a polyspecific ulins and/or complement components. Hemolytic
AHG, it is advisable to perform further tests with anemia may or may not be present (see later).
monospecific reagents in all patients who have a pos- 2. Antibodies against drugs attached to RBC membrane
itive DAT and who have hemolytic anemia. This is (e.g., penicillin) (see Chapter 8).
true because such testing provides additional infor- 3. RBC-bound immune complexes, or complement bound
mation that is helpful in determining the specific type to “innocent bystander” RBCs by complement activa-
of immune hemolytic anemia that is present.17,18 For tion remote from RBCs (e.g., anti–drug-drug complex)
this purpose, the most important reagents are a (see Chapters 8 and 9).
monospecific anti-IgG and an anti-C3 containing 4. Nonspecific uptake of protein: From 1% to 15% of
anti-C3d/C3dg activity (which may contain anti- random hospital patients and 0.01% to 0.1% of
bodies against other C3 determinants).7 Such antisera, blood donors have been reported to have positive
licensed in the United States by the Food and Drug DATs.20,21 Many of these positive DATs are
Administration (FDA) for use in the DAT, are readily thought to be due to nonspecific uptake of IgG
available. from the plasma (cytophilic IgG). Several reports
In the unusual instance in which a patient is sus- indicate that 65% to 80% of RBCs with positive
pected of having an immune hemolytic anemia but DATs yield nonreactive eluates, suggesting that
has a negative DAT with AGS containing anti-IgG and no RBC antibodies are present on the cell. Toy
anti-C3d, the use of anti-IgM and anti-IgA antibodies and coworkers22 and Heddle and coworkers23
may be indicated (see Chapter 9). At the present, anti- showed a relationship between positive DATs
sera of such specificities have not been licensed in the yielding nonreactive eluates and elevated plasma
United States for use as AGS (i.e., for use with RBCs), gamma globulin levels. Sometimes drugs can
but antisera of these specificities that are manu- modify the RBC membrane, leading to the
204 Immune Hemolytic Anemias

adsorption of many proteins, including IgG. An c. Resuspending centrifuged RBCs too vigorously
example of this is the positive DAT sometimes may disperse weak agglutination.
associated with cephalosporin administration (see 2. AGS does not contain appropriate antibody: As
Chapter 8). It is also possible that this is the cause mentioned elsewhere, commercial polyspecific
of the positive DAT that have been described AHG may have insufficient anti-C3d,7,8,11 anti-
associated with some bacterial or viral infections. IgA,16,30 or anti-IgM11,16 to detect these proteins
5. Hemolytic transfusion reactions (HTRs): Delayed sensitizing a patient’s RBCs.
HTRs can often be very difficult to differentiate 3. Low-affinity antibody sensitizing a patient’s RBCs.
serologically from AIHA (see Chapter 9). Allo- We occasionally encounter patients with RBCs sen-
antibodies present in the recipient can sensitize the sitized with autoantibody that does not seem to
transfused donor RBCs, leading to a positive DAT. “fit” very well; that is, it dissociates very easily,
If the alloantibody(ies) present in the plasma of the even during routine washing of the RBCs for per-
donor react with most RBCs (e.g., antibody to a formance of the DAT. It is important to add the
high frequency antigen or a mixture of antibody AHG to the washed RBCs immediately after
specificities), they can appear to be autoantibody. washing and to read the AGTs immediately after
6. Passive transfer of alloantibody in transfused prod- centrifugation, as these autoantibodies will elute
ucts24: Alloantibodies may be present in plasma from the RBCs very rapidly after washing. More of
products (e.g., platelets, IVIg), or even donor the autoantibody is lost from the RBCs when they
RBCs. Such antibodies (including ABO antibod- are washed at 37°C than at room temperature or
ies) can sensitize the recipient’s RBCs and some- below, even though the patient has warm-type
times cause an HTR. AIHA. If we suspect this is occurring, we wash the
7. Hemolytic disease of the newborn: Maternal antibod- RBCs in ice-cold low ionic strength or normal ionic
ies can cross the placenta and sensitize fetal RBCs strength saline with appropriate controls (e.g., after
(see Chapter 13). washing, centrifuge 1 vol of 2% to 5% washed RBCs
8. High reticulocyte count: There have been reports in 2 vol AHG diluent [if available] or 6% albumin in
indicating that high reticulocyte counts cause a separate “control” tube, to ensure that the cells
false-positive DATs due to anti-transferrin in the are not agglutinated due to cold autoantibodies).16
AHG.25,26 We have not found this to be a problem We have encountered several patients with acute
with modern polyclonal reagents, and this will AIHA whose DATs were negative when the cells
not occur with monoclonal AGS. Similarly, were washed at 37°C, 1+ when washed at room
Chaplin18 and Worlledge27 have not found this to temperature, and 3+ when washed in ice cold
be a cause of false-positive results in their series. saline. It has been suggested that IgG serum allo-
9. Polyagglutinable RBCs: When the T antigen is antibodies and autoantibodies that agglutinate only
exposed on the RBC membrane (e.g., by bacterial enzyme-treated RBCs are of such low affinity that
infection), a false-positive DAT can occur if they wash off untreated RBCs during the washing
sufficient anti-T is present in polyclonal (e.g., process of the IAT.31 Such antibodies may be
rabbit) AHG. We have not observed this in com- similar in nature to the autoantibodies discussed
mercially available polyclonal AHG, probably earlier that have sensitized the patient’s RBCs in
because the rabbit serum is diluted considerably vivo.
or fractionated in the preparation of the AHG. 4. RBC-bound IgG autoantibody may be present in
Monoclonal AGS will not contain anti-T. too low a concentration to be detected by the AGT.
10. Silica, derived from glass, and metallic ions, have been Before a positive AGT is clearly visible, the RBCs
described as a cause of false positive DAT.28,29 We usually must be sensitized with 150 to 500 mole-
have not encountered any examples of this. cules of IgG per RBC.32-35 Sometimes, more sensi-
tive techniques will show IgG to be present on the
RBC (see Chapter 9 for a full discussion).
Causes of False-Negative DATs
1. Poor technique
a. Insufficient washing of RBCs will enable resi- IgG and Complement on RBCs
dual plasma globulin to inhibit the AHG. To of DAT-Negative Healthy Individuals
control this, known IgG weakly sensitized RBCs RBC-BOUND IgG
must be added to every negative test and
recorded as positive (i.e., indicating uninhibited Gilliland and coworkers34 and Petz and Garratty,35
AGS to be present in the tube) before the using a complement fixation antiglobulin consump-
negative test can be accepted as truly negative. tion assay, found 35 or fewer IgG molecules per RBC
b. Use of saline or AGS contaminated with human when testing DAT-negative healthy individuals.
globulin (1 part in 4000 parts of normal plasma/ Investigators in England and Canada came to very
serum is often sufficient to completely inhibit similar conclusions using radiolabeled anti-IgG to
the anti-IgG in an AHG reagent; this can be measure IgG on the RBCs of healthy individuals.
controlled as given in a). Merry and coworkers36 found the RBC to have 5 to 90
The Serologic Investigation of Autoimmune Hemolytic Anemia 205

(mean, 39) IgG molecules per RBC, and Jeje and tion may be involved. Lutz and coworkers43 suggested
coworkers37 found 3.7 to 16 fg of IgG/103 RBCs, with that the antigen is a dimer of band 3. Low and cowork-
a mean of 14 fg/103 RBCs, or 31 IgG molecules per ers44 presented evidence that as hemoglobin begins to
RBC. Thus, there seems to be no doubt that normal denature, it forms hemichromes that crosslink band 3
healthy individuals have IgG on their RBCs, but it is into clusters, and suggested that it is these clusters that
still not clear whether it is present on all or only some provide the recognition site for antibodies directed
RBCs, whether the amount varies a great deal from against the senescent cell antigen.
cell to cell, and whether it serves a physiologic role.
The source of RBC-bound IgG in healthy individu- RBC-BOUND COMPLEMENT
als is not clear. Is the IgG representing IgG RBC
autoantibody or nonspecifically bound IgG (“cyto- Complement also has been detected on the RBCs of
philic IgG”), or are both populations present? If either healthy individuals. Using potent anti-C3d reagents
or both are present, are they present on all RBCs or and the standard AGT, Graham and coworkers45
only on a subpopulation (e.g., older RBCs)? showed that RBCs from most individuals were coated
Kay38 found that RBCs aged in vitro were phagocy- with C3d; other workers have confirmed this. Using the
tosed when they were incubated in autologous or allo- AutoAnalyzer, Rosenfield and Jagathambal46 reported
geneic IgG and then incubated with autologous 5 to 40 C3 molecules per RBC. Using radiolabeled anti-
macrophages. In vitro aged RBCs incubated in IgM-, C3, Freedman and Barefoot47 reported 207 to 427 C3d
IgA-, and IgG-depleted serum or medium were sepa- molecules per RBC; Chaplin and coworkers48 reported
rated, by density, from freshly drawn human blood. 50 to 160 C3 molecules per RBC, and Merry and
Less than 5% of the young cells were phagocytosed, coworkers49 found 280 to 560 C3d molecules per RBC.
whereas more than 30% of the old cells were phago- The differences among these three reports perhaps
cytosed, independent of the incubation medium. In reflect the specificity of the anti-C3 used (e.g., the pro-
further studies, Kay eluted the immunoglobulin from portion of antibody activity directed against various
the older RBCs and showed that it was IgG1 and IgG3. epitopes, such as C3d or C3dg, on the RBC-bound C3).
The eluted IgG would reattach only to stored autolo- Freedman50 showed that older RBCs, separated
gous or allogeneic RBCs; after treatment, these cells from fresh blood, had more C3d on their membrane
were phagocytosed by autologous macrophages. No (as well as IgG, IgM, and IgA) than did younger RBCs;
phagocytosis occurred with young RBCs incubated in they found no increase in C3b, C4d, C5, or factor B.
the eluted IgG. Neuraminidase-treated RBCs incu- They also quoted unpublished observations that
bated in medium were not phagocytosed, but after C3b-coated RBCs have 10% to 15% less free sialic acid
incubation in eluted IgG, they were phagocytosed. than uncoated RBCs and suggested that the accumu-
Finally, Kay and coworkers showed that the phenom- lation of C3 on older RBCs may relate to their removal
enon was not due to nonspecific binding of IgG, from the circulation. They did not believe that the
because adsorption of pooled normal IgG with aged complement was on the RBCs as a result of comple-
RBCs abolished its phagocytosis-inducing activity; ment activation by IgG autoantibody (i.e., senescent
receptor blockade studies showed that the IgG was cell antibody), as RBC-bound C4 was not increased on
attaching to the RBCs by its Fab portion. The authors older cells. This would suggest complement activa-
suggested that the cell-bound IgG was a physiologic tion by the alternative pathway rather than antibody-
autoantibody contributing to the maintenance of mediated classic pathway activation. RBC-bound
homeostasis by collaborating with macrophages in complement has been shown to accumulate on RBCs
the removal of senescent and damaged cells. stored in vitro. It is of some interest that Szymanski
Several other publications support the hypothesis of and coworkers51 found that the 24-hour survival of
Kay and coworkers. Alderman and coworkers39 stored RBCs showed a negative correlation with the
reported that 85% to 95% of older RBCs had mem- length of storage and the amount of RBC-bound C3.
brane-bound IgG, whereas no IgG was detected on the They suggested that RBC-bound C3 may contribute
younger RBCs. They confirmed the findings of Kay and significantly to the storage lesion of stored RBCs.
coworkers that the IgG eluted from older RBCs would There are many reasons why RBCs may have small
combine in vitro with older RBCs but not young cells. amounts of complement on their surface, other than as
They also demonstrated that the eluted IgG would a result of complement activation by RBC autoantibod-
bind to neuraminidase-treated RBCs, suggesting that ies and alloantibodies. One reason may relate to the
the antigenic site revealed during enzyme treatment in presence of complement receptor (CR1) on RBCs. This
vitro is similar to that developing during aging in vivo. receptor can bind C3b, iC3b, and C4b. Although all of
Szymanski and coworkers40 used a sensitive auto- these complement molecules are activation products
mated AGT to confirm the presence of IgG on normal and might not be expected to be present in healthy indi-
RBCs. They showed that separated young RBCs had viduals, there is increasing evidence that low-level acti-
less membrane-bound IgG than older cells. Freedman41 vation may be occurring continually, and small
had similar findings using 125I-labeled anti-IgG. Kay42 amounts of C3b and its breakdown products, iC3b and
suggested that the senescent cell antigen is derived C3d,g, may be commonly present in the plasma. It is
from band 3, probably via degradation, and that oxida- possible that some of these activation products may
206 Immune Hemolytic Anemias

bind to the CR1 receptor of RBCs, accounting for a because sometimes the results in the reports from the
small number of “naturally occurring” RBC-bound United Kingdom related to DATs performed on
complement molecules. It has also been shown that tiles and read macroscopically; the DAT results in Los
RBCs can be involved as “innocent bystanders” in com- Angeles were from a “spin” tube technique, where all
plement activation remote from the RBC. This might macroscopic negatives were checked microscopically.
occur in infections or immune complex formation (e.g., Approximately 18% of the DAT reactions were graded
in systemic lupus erythematosus). For instance, Salama as 2+, and 3% were graded as 3+ or higher. Sixty-seven
and Mueller-Eckhardt52 showed that complement acti- percent of the positive DATs were associated with IgG
vation by bacteria could lead to complement sensitiza- sensitization, 32% had RBC-bound IgG but no comple-
tion of nonsensitized bystander RBCs. It follows that ment, 35% had RBC-bound IgG and complement, and
one might expect to find more RBC-bound complement 33% had only RBC-bound complement.
in sick patients than in healthy individuals, and there is Two English studies suggest that it is rare for a
good evidence for this (see later). Siegel and cowork- positive DAT in blood donors to be associated with
ers53 suggested that RBCs were perhaps more impor- hemolytic anemia. Gorst and coworkers56 recalled 32
tant than macrophages in clearing immune complexes donors who had a positive DAT and studied them in
from the circulation. They suggested that the main detail. Twenty-six (81%) still had a positive DAT 6
function of the RBC CR1 receptor may be to attract months up to 18 years after the initial finding.
complement-containing immune complexes, leading to Seventeen (53%) of the 32 were associated with IgG
macrophage interaction and subsequent clearance. sensitization; of these, 13 (41%) of the 32 donors still
Thus, it is easy to accept that RBCs may be coated with had IgG-sensitized RBCs when retested. They found a
small amounts of complement independent of activa- strong correlation between increasing age of donors
tion by antibody directed against RBC antigens. and positive DATs. No clinical abnormalities were
detected in any of the 32 donors. All hematologic and
Seemingly Healthy Individuals biochemical tests, including reticulocyte counts, were
with Positive DATs Due to IgG normal. One donor, after having a positive DAT for
and/or C3 Sensitization 2 years, suddenly developed WAIHA that required
splenectomy for its control. In a similar study, Bareford
It has been known for many years that some apparently and coworkers58 also recalled 26 DAT-positive donors
normal individuals have positive DATs, but the inci- for study. All of the positive DATs were associated with
dence among normal blood donors varies somewhat in IgG sensitization. Of these donors, 9 (35%) still had a
the literature.54-58 Using an AGS that contained mainly positive DAT 1 to 18 years after the original finding.
anti-IgG, Weiner54 reported 18 cases in a British donor One of the donors had developed AIHA soon after the
population of 60,000 (1:3300). Most of the cases initial positive DAT 10 years earlier and required treat-
reported by Weiner had strongly positive (>2+) DATs. ment with steroids and splenectomy. One donor had
Habibi and coworkers55 reported an incidence of developed ulcerative colitis, one had mild arthritis, and
1:13,000 in a large French donor population; 97% of the other five remained in good health. As in the series
their positive DATs were associated with IgG sensitiza- of Gorst and coworkers,56 an increasing incidence of
tion. Gorst and coworkers56 reported 65 donors with positive DATs was noted with increasing age.
positive DATs encountered in a 14-year period (an inci- Habibi and coworkers55 followed 63 French DAT-
dence of approximately 1:14,000 donations, but their positive donors for 5 years and, in contrast to the UK
figures were not corrected for the incidence in donors, studies, found 72% of them to have some evidence of
rather than units donated); approximately 50% of these increased RBC destruction as evidenced by increased
positive DATs were associated with IgG sensitization reticulocytes, hyperbilirubinemia, and abnormal T50Cr
(23% of these had complement in addition to IgG of labeled RBCs (Cr studies were performed on only 12
detectable on the RBCs). Bareford and coworkers58 of the DAT-positive donors). These 63 were selected
reported an incidence of positive DATs in their donor from 69 DAT-positive donors detected in 892,000
population in Leeds (United Kingdom) over a 20-year donors (an incidence of 1:13,000). Almost all (97%) of
period as 1:7500. All of the DAT-positive donors tested the positive DATs were associated with IgG sensitiza-
had IgG on their RBCs, 12% of these had RBC-bound tion. Seventeen (25%) were due to methyldopa therapy.
complement in addition to IgG. Only 10% of the donors had a mild anemia (hemoglo-
Allen and Garratty57 found 1:1000 of 1.3 million bin <13 g/dL in men and <12 g/dL in women).
donors in Los Angeles, tested over a 5-year period, had Seventeen percent had an increased reticulocyte count,
a positive DAT. Eighty percent of the DAT reactions and 12% had hyperbilirubinemia in addition to
were 1+ or less (50% were <1+). The difference in increased reticulocytes. RBC survival studies were per-
reading or recording very weakly positive DATs may formed on cells of 12 donors, using autologous 51Cr-
account for the higher incidence reported from Los labeled RBCs, and 61% were found to be abnormal.
Angeles compared with other areas (Table 6-2 shows Twenty-eight percent of the donors had a T50Cr of more
agglutination reactions and scores as used by our labo- than 26 days, and 33% had a T50Cr of less than 24 days
ratory at the time most of the data in this book were (normal T50Cr, 30 ± 3 days). These figures suggest that
gathered). This may be particularly true for Europe, somewhere between 1:18,000 and 1:21,000 (61% to 72%
where the reported incidences were much lower, of all DAT-positive donors) of French blood donors
The Serologic Investigation of Autoimmune Hemolytic Anemia 207

TABLE 6-2. AGGLUTINATION GRADING AND SCORES

Grading Appearance of Red Cells Score

++++ or 4+ One large agglutinate; no unagglutinated cells 10


+++ or 3+ Several large agglutinates, few free cells 8
++ or 2+ Large agglutinates in a sea of smaller clumps and some free cells 6
+ or 1+ Many small agglutinates (approximately 20 RBCs per agglutinate) 4
(+) or (1+) or 1⁄2+ Scattered small agglutinates in a sea of free cells. Just visible macroscopically 3
± or micro Small agglutinates visible only microscopically 1
0 No agglutinates seen 0

present with some signs of AIHA. These findings can be The highest incidence was reported by Judd and
contrasted to the findings in England of approximately coworkers,63 who found 15% of patients undergoing
1 case of AIHA per 1 million donations (not donors) pretransfusion tests to have a positive DAT; 55% of
reported by Gorst and coworkers56 and 1 case per 5 these (8% of a total of 65,000 patients) were associated
million donors studied by Bareford and coworkers.58 with IgG sensitization. In a later paper from the same
Pirofsky59 reported the incidence of AIHA in the United hospital, Judd and coworkers64 reported a slightly
States as approximately 1:80,000 of the general popula- lower incidence (6%) of pretransfusion patients with
tion, which may be a higher incidence than a typical positive DATs associated with IgG sensitization. Huh
blood donor population, where most donors are 17 to and coworkers65 found 3.5% of pretransfusion
70 years old. It should be emphasized that Pirofsky’s samples, or 10% of patients (14,548 pretransfusion
incidence only relates to patients with clinically obvious blood samples from 6959 patients), had a positive
hemolytic anemia, in contrast to the French study DAT; 68% of these were associated with IgG sensitiza-
where individuals with an abnormal laboratory test tion (42% IgG only and 26% IgG plus C3), 19% had C3
result (DAT) were carefully studied for any signs of only on the RBCs, and 14% were said to be equivocal.
increased RBC destruction (e.g., use of 51Cr-labeled
RBCs). Thus, it would seem that increased RBC destruc- Clinical Significance of Positive DATs
tion due to IgG autoantibodies, in blood donors, may be in Patients
more common than is generally assumed, but many of
these have a relatively normal hemoglobin and proba- It is difficult to assess the clinical significance of a
bly never feel sick enough to go to a physician. positive DAT in random patients. If one relates clinical
significance only to RBC survival, then a positive DAT
associated with IgG is probably more significant than
Positive DATs in Hospitalized Patients one due to only complement sensitization, but in many
Many more patients than donors have positive DATs patients it may have no significance (e.g., see Toy and
but may also not have hemolytic anemia. Once again, coworkers22 and Heddle and coworkers,23 discussed
there is considerable variation in incidence, even in later). Bohnen and coworkers61 found that 60% of their
accounts from the United States, where the reported DAT-positive patients had signs of anemia but found it
incidence varies from 1% to 15%.20,21 It is not clear difficult to explain the etiology of the anemia.
why reports vary so much. Different types of AGS Reticulocytes were raised above 10% in only 17% of the
(anti-IgG versus polyspecific) account for some varia- anemic patients, and above 5% in 31%; these results
tions, but the major factor may be the types of patients included defined cases of AIHA. Very little evidence
predominating in certain medical centers and the was presented for hemolysis in most of the anemic
number of patients tested. patients. Of the patients with positive DATs, 39% had
Using AGS containing powerful anti-C3d activity, received blood transfusions during the previous
Worlledge and Blajchman,14 Petz and Garratty,35 2 months, and alloantibodies were detected in 25% of
Freedman,60 and Chaplin and coworkers48 all these patients. Eluates were not tested, but it is proba-
reported a 7% to 8% incidence of positive DATs in hos- ble that many of the DATs in this recently transfused
pital patients. More than 80% of these were due to population were due to alloantibodies or would have
complement sensitization, with no IgG detectable. nonreactive eluates (see later).
Bohnen and coworkers61 found an incidence of 1% Toy and coworkers66 reported that 18% of patients
positive DATs in a 13-year study of hospitalized with acquired immune deficiency syndrome (AIDS)
patients, where 17,110 DATs were performed. Lau and had a positive DAT. This contrasted with an incidence
coworkers62 found a 0.9% incidence of positive DATs of only 0.6% positive DATs in their hospital population.
when testing 6883 medical and surgical admissions to Of the positive DATs in AIDS patients, 80% were asso-
the hospital. Lau and coworkers62 used only an anti- ciated with IgG sensitization of RBCs. Although all
IgG reagent, and it is probable that when Bohnen and AIDS patients were anemic, no obvious evidence of
coworkers61 performed their study (1950 to 1963), the clinical hemolysis was noted, but no 51Cr-labeld RBC
commercial AGS was predominantly anti-IgG. survival studies were performed. Because serum
208 Immune Hemolytic Anemias

globulin levels were raised in the DAT-positive AIDS used for the DAT; and (3) low-affinity IgG autoanti-
patients and eluates from the patients’ RBCs were non- bodies, which are washed off the RBCs during the
reactive, Toy and coworkers66 suggested that the posi- washing phase of the DAT (see Chapter 9).
tive DATs might be due to nonimmune adsorption of
IgG onto the patient’s RBCs. In a separate study, Toy
and coworkers22 found that elevated serum globulin DETECTING SMALL AMOUNTS OF
and blood urea nitrogen levels correlated significantly RBC-BOUND IgG (AND IgA AND IgM)
with positive DATs associated with nonreactive RBC
eluates in patients who did not have AIDS. They found
0.7% of their hospital patients to have a positive DAT Using Flow Cytometry69
associated with IgG sensitization, and 79% of these 1. Wash test RBCs and control RBCs four times
RBCs yielded a nonreactive eluate. Seventy-five percent with 4°C phosphate-buffered saline (PBS).
of these cases had elevated serum globulins, in contrast Suspend to 3% to 5% with 4°C PBS. Perform all
to 29% of DAT-negative controls. The major causes of of the following steps in cold (cold reagents, cold
elevated serum globulins were liver disease and infec- washes, cold incubations in refrigerator or dish-
tion. Heddle and coworkers23 have also described a pan with ice water) as much as possible.
correlation with hypergammaglobulinemia and RBC- 2. For each RBC sample to be tested, label at least
bound IgG. Fifteen DAT-positive patients with nonreac- two tubes. Label one tube as a control, and the
tive eluates had elevated IgG levels (range, 1450 to 7730 other(s) with the antisera to be tested (e.g., anti-
mg/dL; mean, 2621 mg/dL). A prospective study of 44 IgG, anti-IgM, etc.).
patients with elevated serum IgG levels yielded three 3. Place 0.1 mL of 3% to 5% washed RBCs in each
positive DATs and these occurred in patients with the tube.
highest IgG levels (2381, 3441, and 3520 mg/dL). 4. Add 0.01 mL of the appropriate dilution of
These findings are extremely important because they fluorochrome-labeled antisera to the appropri-
suggest that in a hospital population, most (about 65% ately labeled tubes.
to 80%) of the positive DATs due to IgG sensitization are 5. Add nothing to the control tubes (background).
due to nonimmunologic adsorption of plasma IgG and 6. Vortex all tubes.
are of no clinical significance. Thus, approximately only 7. Incubate 30 minutes (e.g., room temperature
1 in 125 hospital patients have IgG-positive DATs with for fluorescein isothiocyanate (FITC)-labeled
any potential clinical significance.20 Positive DATs of antibodies, 4°C for phycoerythin (PE)-labeled
potential significance include IgG autoantibodies, drug- antibodies).
induced antibodies, and alloantibodies that may or may 8. Wash RBCs once with 0.2% BSA/PBS for FITC-
not cause increased RBC destruction. labeled antibodies or 2% bovine serum albumin
Using radiolabeled anti-C3d, Chaplin and cowork- (BSA)/PBS for PE-labeled antibodies.
ers67 and Freedman and coworkers68 showed that 9. Add 0.1 mL PBS to cell button and mix by
random hospitalized patients had more RBC-bound drawing RBCs up and down, using a small-bore
C3 than did healthy individuals. Chaplin and cowork- pipette (to break up any small agglutinates that
ers67 found that 33% of 313 patients had levels of RBC- may be present).
bound C3d above the normal range; only 8% of the 10. Add a subsample of each RBC sample to a 12 ×
patients had enough RBC-bound C3 to be detected by 75-mm polystyrene tube containing 1 to 2 mL PBS
the DAT. The great majority of the patients with so that an RBC suspension of approx-imately
increased RBC-bound C3d did not have any known 0.2% is obtained (flow rate on the flow cytometer
autoimmune disease. Freedman and coworkers68 should be around 500 to 1000 events per second).
studied 227 hospitalized patients; 72 (32%) were 11. Immediately before aspiration into the flow
found to have moderately increased levels of RBC- cytometer, mix RBCs by vortexing.
bound C3d. In contrast to Chaplin and coworkers,67 12. Acquire at least 10,000 events for each sample.
Freedman and coworkers68 found that these increases 13. Analyze data. Place electronic region around the
were more often found in patients with conditions RBC population based on forward versus side
where complement activation might be expected. scatter cytogram of 1 normal RBC control.
Twenty-six (13%) of 203 patients had markedly ele- a. Printout forward versus side scatter cytogram
vated RBC-bound C3d; these patients (>1100 mole- and fluorescence histogram for each sample.
cules C3d per RBC) almost always had an associated b. Record median relative fluorescence intensity
autoimmune disease (e.g., AIHA). (RI):

RI = Median fluoresence of RBCs ±


AIHA Associated with a Negative DAT fluorochrome-labeled antibody
The most common causes for this are (1) RBC-bound Median fluorescence of RBC background control
IgG below the threshold of the AGT (i.e., <200 IgG
molecules per RBC); (2) RBC-bound IgA and IgM, c. Overlay RBC background control fluorescence
which are not detectable by most routine reagents histogram on top of RBCs + fluorochrome
The Serologic Investigation of Autoimmune Hemolytic Anemia 209

antibody histogram. Use subtract graphs func- Materials


tion. Divide number of events after subtraction 0.9% NaCl, unbuffered
by number of gated events in RBCs plus fluo- 9 g NaCl
rochrome antibody. Record this as percent 1 L deionized H2O
positive result for all samples. Store at room temperature
d. Determine mean + 2 SDs for median RI and LIM (low ionic medium)
percent positive for all the control results. For 100 mL
5 g dextrose
Quality Control 0.2 g Na2EDTA
1. RBCs plus nothing (background) control gives Bring to 100 mL with deionized H2O
an indication of autofluorescence background. Store at 2°C to 8°C
The patient’s RBC autofluorescence may be Polybrene (hexadimethrine bromide, a very hygro-
increased (compared with the normal donor’s scopic polymer) has to be stored in a desiccator
RBCs) due to medications, etc. If so, then inter- at (2°C to 8°C)
pretation of results with fluorochrome-labeled 10% Stock solution:
antibodies may be difficult. 1 g hexadimethrine bromide
2. To control for RBC agglutination (e.g., when Bring to 10 mL with 0.9% unbuffered NaCl
testing anti-IgM), test another antiserum ex- Store in a plastic container at 2°C to 8°C
pected to be nonreactive (e.g., anti-IgG, anti-IgA) Working solution (0.05%, or 1:200 dilution of the 10%
in parallel. If anti-IgG or anti-IgA is reactive, this stock):
may indicate the presence of that immunoglobu- 0.05 Polybrene stock + 9.95 mL unbuffered NaCl
lin on those RBCs. If both anti-IgG and anti-IgA Resuspending solution
are reactive, that may indicate that RBC agglu- 3.35 g trisodium citrate (Na3C6H5O7)•2H2O
tination is present. The anti-IgG and anti-IgA 2 g dextrose
results may then be used as background to Bring to 100 mL with deionized H2O
determine if the anti-IgM results are positive. Store at 2°C to 8°C
PBS, pH 7.3
Interpretation AB plasma
1. Patient’s RBCs with median RI and percent pos- 5% AB plasma in PBS (v/v)
itive results of greater than mean + 2 SDs of 2 mL AB plasma
normal controls are positive for the relevant 38 mL PBS
immunoglobulin. Store at −20°C in 0.5-mL aliquots
2. Patient’s RBCs with only median RI or percent 12 × 75-mm test tubes
positive results of greater than mean + 2 SDs Uniform drop pipettes
need to be evaluated individually (e.g., visual D+ RBCs for the controls, 3% to 5% (v/v)
inspection of histogram). Commercial anti-D diluted in 5% AB plasma so
3. When testing RBCs with cold or spontaneous that it does not react by routine tube indirect
agglutination, compare results, for example, of AGT (e.g., 1:8000). Store frozen at −20°C in 0.5-mL
patient’s RBCs plus fluorochrome labeled anti- aliquots
IgM versus patient’s RBCs plus fluorochrome Anti-IgG
labeled antisera expected to be nonreactive. Antiglobulin control cells

Procedure
1. Label 4 12 × 75-mm tubes for patient’s washed
DIRECT POLYBRENE TEST RBCs, patient’s unwashed RBCs, the positive
control, and the negative control.
2. Add 2 drops of 5% AB plasma and 1 drop of
Principle the patient’s washed 3% to 5% RBCs to the
Polybrene (Sigma, St. Louis, MO) is a quarternary “washed” tube.
ammonium polymer (hexadimethrine bromide) that 3. Add 3 drops of a 1.5% suspension of the
facilitates detection of RBC antibodies. The RBC sen- patient’s unwashed RBCs suspended in auto-
sitization phase occurs in a low ionic medium (LIM). logous plasma to the “unwashed” tube.
Polybrene is then added to nonspecifically aggregate 4. Add 2 drops of the diluted anti-D and 1 drop of
the RBCs, which permits crosslinking of antibody D+ RBCs to the positive control tube.
molecules through close approximation of the RBCs. 5. Add 2 drops of 5% AB plasma and 1 drop of D+
Hypertonic salt solutions, such as sodium citrate, dis- RBCs to the negative control tube.
perse nonspecific aggregation of the RBCs, but anti- 6. To all tubes add 1 mL of LIM. Mix.
body-mediated agglutination of IgG-coated RBCs 7. Incubate 1 minute at room temperature.
will persist. The test can then be converted to an AGT 8. Add 2 drops of working Polybrene solution to
if antibody is not detected by the Polybrene effect. all tubes and mix.
210 Immune Hemolytic Anemias

9. After 15 seconds, centrifuge at 1000 × g for sometimes contain heterophil antibodies that have to
10 seconds. be removed by dilution or adsorption with non-
10. Decant supernatant. sensitized human RBCs). The quality control must
11. Add 2 drops of resuspending solution. be precise, as agglutination is a far more sensitive
12. Mix gently (shake rack at 45-degree angle) for technique than some immunologic techniques.
10 seconds. Monospecificity by some immunologic techniques
13. Read macroscopically and microscopically (e.g., precipitation or flow cytometry) does not ensure
within 3 to 5 minutes. Do not recentrifuge. monospecificity by the AGT (e.g., an anti-IgA that
14. Add 1 drop of resuspending solution. Mix. does not show any precipitin line against IgG may
15. Add 2 drops normal (neat) AB plasma. Mix. react 4+ with IgG Rh antibody sensitized RBCs). Thus,
16. Wash 3 times with PBS. anti-IgA must always be shown to be nonreactive by
17. Add anti-IgG, centrifuge, and read. Read all the AGT against RBCs strongly sensitized with IgG,
macroscopically negative results under the mic- IgM, and complement; similarly, anti-IgM must not
roscope. Record results. react with IgG, IgA or complement sensitized RBCs.
18. Add antiglobulin control cells to all negative
tests, centrifuge, read, and record macroscopi-
cally positive results. Standardization of anti-IgA and anti-IgM. It is
difficult to find pure nonagglutinating IgA and IgM
Interpretation blood group antibodies to prepare IgA/IgM-coated
1. The presence of RBC-bound antibody is recog- RBCs; therefore, we standardize our AHG using a
nized by the persistence of agglutination after the passive agglutination technique. We coat chromic
Polybrene effect has been neutralized (resuspen- chloride-treated RBCs with purified IgA or IgM pro-
sion phase) and/or by reactivity with anti-IgG. teins and then test these coated RBCs by the regular
2. Negative tests indicate the absence of RBC- AGT.
bound IgG, provided that the positive control
was reactive.
CHROMIC CHLORIDE METHOD FOR
Notes
COUPLING PROTEINS TO RBCS
1. Compare tests with the negative control when
examining for persistence of agglutination after
adding the resuspending solution. Materials
2. The activity of the Polybrene can vary; the Unbuffered saline (UBS), 0.9% NaCl.
reagent is very hygroscopic and reactivity can PBS, pH 7.3.
weaken over time. Reagents should be checked Chromium chloride, 6-hydrate (CrCl3•6H2O): 1%
when first put into use after preparation with the stock solution prepared in deionized water.
anti-D dilution previously used for verification Store in dark bottle at 2°C to 8°C.
of reactivity. The dilution of the control antibody Antigen or antibody to be coupled to the RBCs (e.g.,
may need to be adjusted with new lots of purified Ig)
Polybrene reagents. Group O RBCs (>24 hours old, <8 days old)
3. Excess citrate can cause dissociation of antibody- Test tubes, glass (never use plastic)
dependent RBC agglutinates. Appropriate antisera to determine if antigen/anti-
4. Read results shortly after Polybrene is neutral- body is bound to RBCs
ized or negative results will occur.
5. Antibodies in the Kell system sometimes fail to Procedure
react or are weaker in the Polybrene system 1. Prepare dilutions of stock CrCl3 in UBS (e.g., 1 in
(resuspension phase). The antiglobulin phase 10, 1 in 20, 1 in 40).
should enable detection of these antibodies. 2. Prepare dilutions of antigen or antibody in UBS
6. Antiglobulin reagents containing anti-comple- (e.g., 0.5, 1, 2 mg/mL).
ment should not be used after Polybrene treat- 3. Wash and pack RBCs.
ment because complement can be fixed to RBCs 4. Label several tubes, one for each of the different
in the low ionic phase. combinations of CrCl3 and the antigen or anti-
body to be used (i.e., a checkerboard titration).
Also include a tube (for each CrCl3 dilution)
where no antigen/antibody is added, just UBS +
DETECTING RBC-BOUND IgA AND IgM
RBCs + CrCl3.
5. In the labeled glass test tubes, place 1 vol (e.g.,
At present, in the United States, anti-IgA and anti-IgM 0.1 mL) antigen or antibody solution and 1 vol
are not available as licensed reagents for use with packed washed RBCs.
RBCs. They are readily available as immunologic 6. Add 1 vol of diluted CrCl3, mixing well during
reagents and can often be used as long as they are this addition.
carefully standardized and controlled (e.g., they 7. Mix well for 5 minutes at room temperature.
The Serologic Investigation of Autoimmune Hemolytic Anemia 211

8. Wash RBCs four times with UBS (do not cen- ii. Add 1 mL of whole blood. This can be fresh
trifuge longer than 30 seconds at 1000 × g). blood or ACD or CDP blood that has been
9. Resuspend RBCs to 3% to 5% in PBS. stored at 4°C for less than 3 weeks.
10. Test RBCs against appropriate antibody (e.g., iii. Mix and incubate at 37°C for 15 minutes.
anti-IgA) and 6% albumin. iv. Wash 4 times.
v. Resuspend RBCs to 2% to 5%.
Quality Control b. Using nonagglutinating Lewis antibodies (e.g.,
Reactivity of the antigen or antibody/CrCl3-treated anti-Lea, anti-Leb, or anti-Lea + anti-Leb) (the
RBCs with specific antibody and not with 6% amount of complement on the RBCs can be
albumin indicates that the antigen or antibody is varied by using dilutions of the Lewis anti-
coupled to the RBCs. Repeat the CrCl3 treatment bodies).71 If the Lewis antibodies have been
with different amounts of antigen/antibody and/or stored for longer than 48 hours at 4°C or
CrCl3 if these results are not obtained. 1 month at –20°C, then the two-stage EDTA
AGT described below is optimal but not essen-
Interpretation tial. Freshly collected Lewis antibodies (or
A checkerboard titration is used to determine the Lewis antibodies stored below –50°C) can be
optimal combination of antigen or antibody and used with a routine one-stage indirect AGT
CrCl3 concentrations. In general, the higher the Cr (e.g., do not add EDTA and do not perform
ion concentration, the more efficient the coupling, steps v, vi, and vii).
but the more likely it is to also encounter nonspecific i. To 9 vol of Lewis antibody, add 1 vol of
aggregation. The amount of CrCl3 that causes aggre- neutral EDTA (4.45% K2 EDTA•2H2O +
gation depends on the protein concentration. The 0.3% NaOH).
optimal amount of CrCl3 is thus a little below that ii. Add 1 vol of 50% washed Lewis-positive
which causes aggregation. RBCs.
Choose the combination of antigen or anti- iii. Incubate at 37°C for 30 to 60 minutes.
body/CrCl3 that gives no aggregation of the RBCs iv. Wash 4 times.
tested with 6% albumin and gives the desired v. To button of RBCs, add 5 vol of fresh
strength of reactivity with the appropriate antisera. normal serum as a source of complement.
vi. Reincubate at 37°C for 15 minutes.
Notes vii. Wash 4 times.
1. The presence of phosphate ions must be avoided viii. Resuspend RBCs to 2% to 5%.
during the coupling procedure, because they c. Using anti-I (the amount of complement on the
will result in the precipitation of chromic ions. RBCs can be varied by using dilutions of the
2. The source of CrCl3•6H2O can be important. anti-I).71
3. Aged CrCl3 (3 weeks) is more efficient in cou- i. Titrate an anti-I and select a dilution that
pling than freshly prepared solutions. To ensure agglutinates OI adult RBCs approximately
even coupling, the CrCl3 should be added drop- 2+ at 20°C to 25°C. This should yield mod-
wise, with constant mixing. erately strongly sensitized RBCs.
4. The order of addition of reactants is important. ii. Dilute the anti-I appropriately in a source
The CrCl3 must be added last, as it is rapidly of complement (i.e., “fresh” inert human
inactivated by protein. serum). For instance, for serum from a
patient with a cold agglutinin titer at 4°C
of 4000; we dilute this anti-I 1:1000 and
1:100 to prepare weakly and strongly
C3b/C4b sensitized RBCs, respectively.
STANDARDIZATION OF
iii. Add 0.1 vol of 50% washed RBCs.
ANTICOMPLEMENT AGS
iv. Incubate at 20°C to 25°C for 10 to
15 minutes.
Preparation of RBCs Coated with Various v. Incubate at 37°C for approximately 5 to
Complement Compounds 10 minutes (to allow agglutination to
The methods below are those we have used success- disperse).
fully in our laboratory. Hoppe70 described methods vi. Wash RBCs 4 times at 37°C.
recommended to manufacturers of AGS by the FDA. vii. Resuspend RBCs to 2% to 5%.

1. C3b- and C4b-coated RBCs71


a. Using low ionic strength 2. C3b (but not C4b)-coated RBCs72,73
i. Prepare 10 mL of 10% sucrose in water. Diluent Solution A
(This can be done simply by adding sucrose In a 250-mL flask containing approximately 125 mL
to the 1-mL mark of a 10-mL graduated cen- distilled water, dissolve:
trifuge tube and then adding water to the a. 23.1 g sucrose
10-mL mark.) b. 173 mg NaH2PO4•H2O
212 Immune Hemolytic Anemias

c. 395 mg Na2EDTA•2H2O Daily Quality Control of AGS


q.s. to 250 mL with distilled water For daily quality control, polyspecific AHG need
only be tested against RBCs weakly sensitized with
Diluent: Solution B IgG (e.g., anti-D). Although it could be argued that it
In a 250-mL flask containing approximately 125 mL would be optimal to test the AHG against comple-
distilled water, dissolve ment-sensitized RBCs, it seems unnecessary because
23.1 g sucrose daily quality control is primarily to ensure that the
178 mg Na2HPO4 AGS has not become inactivated by contamination
395 mg Na2EDTA•2H2O or improper storage. There is no evidence to suggest
q.s. to 250 mL with distilled water. that the anticomplement activity can be selectively
Adjust solution A to pH 5.1 by the addition of inactivated under the usual conditions of storage in
solution B (e.g., approximately 6.5 mL). blood banks; in our experience, when any inactiva-
tion occurs (e.g., contamination with human serum),
the anti-IgG reaction will be a sensitive indicator as
0.4 M MgCl2 Additive long as RBCs weakly (approximately 1+) sensitized
To 10 mL distilled water add 810 mg MgCl2•6H2O. with IgG-sensitized RBCs are used as indicator cells;
many commercially available AGT “check cells” are
Coating Procedure too strongly sensitized to show minimal to moderate
a. Whole ACD or CPD blood may be used equally inhibition of the AGS.75 Some commercial package
well. Chill all reactants to ice-water tempera- inserts recommend the use of complement-coated
ture before combining. RBCs, in addition to IgG-sensitized RBCs when
b. Place 19 mL diluent in small flask in ice-water using polyspecific AGS.
bath on magnetic stirrer.
c. With stirrer on, add 1.0 mL ACD or CPD blood 1. Preparation of IgG weakly sensitized RBCs for
dropwise, rapidly. quality control of AGS
d. Immediately add 0.1 mL MgCl2 additive (i.e., a. To each of 12 tubes, add 1 mL of saline.
final concentration of added Mg = 0.002 mg). b. To the first tube, add 1 mL of an IgG anti-D
e. Stir in ice-bath for 1 hour. (e.g., a slide and rapid tube anti-D commercial
f. Wash RBCs 4 times with saline at 0°C. typing reagent), making a 1:2 dilution.
c. Prepare serial doubling dilutions by mixing
3. C4b (but not C3b)-coated RBCs67
saline and antibody, and then transferring
a. Using low ionic strength method
1 mL to the next tube. This is repeated with
i. Add 10 mL of 10% sucrose in water to a
each tube to the 12th tube, and the final l mL
100 X 16-mm tube containing 12 or 15 mg
is discarded.
K EDTA.
d. To each 1 mL of anti-D, add 10 drops of a 5%
ii. Add 1 mL of whole blood as described in
suspension of D-positive RBCs to each tube.
1 (a) (ii) above.
e. Incubate at 37°C for 15 minutes.
iii. Incubate at 37°C for 15 minutes.
f. Centrifuge, remove, and discard supernatant;
iv. Wash 4 times.
wash RBCs 4 times.
v. Resuspend to 2% to 5%.
g. Resuspend RBCs to 5%. Add 1 drop of RBCs
4. C3d/C4d, C3d or C4d-coated RBCs71,74 from each tube to 2 drops of AGS.
a. RBCs sensitized with C3b/C4b, C3b, or C4b h. Centrifuge tubes and inspect for degree of
can be prepared by any of the methods agglutination, carefully noting the strength of
described previously. the reaction (see Table 6-2).
b. An equal volume of packed, washed C3b/C4b, i. Select tubes that show 1+ to 2+ macroscopic
C3b, or C4b-coated RBCs is incubated with agglutination.
0.1% trypsin (pH 7.3) for 30 minutes or inert j. As 1+ sensitized RBCs may appear much
fresh normal EDTA-treated serum (as a source weaker when added to nonsensitized RBCs in
of factors H and I) for 12 hours at 37°C. Non– the “check cell” procedure, it is wise to try
complement-sensitized RBCs should also be several of the sensitized cells (1+ or 2+) as a
incubated with the trypsin, or the normal check cell and choose a dilution that yields
serum, as a negative control. RBCs that will give about 1+ reactions following
c. The RBCs are washed 4 times and resuspended addition to nonsensitized RBCs.
to 2 to 5%. k. A large batch of sensitized RBCs can be pre-
pared, washed, and stored in CPD or Alsevers
Once prepared, any of the above complement-sensi- solution at 4°C; these usually last for up to 1
tized RBCs can be stored frozen in glycerol or liquid month. Alternatively, prepare a quantity of
nitrogen. If the C3b- or C4b-sensitized RBCs are anti-D at this dilution and freeze it in aliquots
frozen immediately after preparation, they do not suitable for use each day.
appear to decay to C3d/C4d on storage.
The Serologic Investigation of Autoimmune Hemolytic Anemia 213

DETECTING LOW-AFFINITY Notes


AUTOANTIBODIES WITH A COLD LOW 1. A column agglutination technique (e.g., gel)
IONIC STRENGTH SALINE WASH DAT16,75,76 anti-IgG DAT may also enhance reactivity of
low-affinity IgG because the patient’s RBCs are
not washed before performing the test.
Materials 2. Washing RBCs at 37°C enhances dissociation of
Test tubes, 10 × 75 mm low-affinity autoantibodies, even when they are
Cold anti-IgG associated with WAIHA.
Cold diluent control (e.g., 6% albumin or manufac-
turer supplied)
Ice-cold low ionic strength saline (LISS) (4°C)
Antiglobulin control cells
Refrigerated centrifuge (e.g., Sorvall RT6000)
STANDARDIZATION OF IgG SUBCLASS
Serologic centrifuge
ANTISERA FOR USE WITH SENSITIZED
RBCs
Procedure
1. Label 2 test tubes: one for anti-IgG, one for control.
2. Add 2 drops of cold anti-IgG and cold diluent The only commercial source of IgG subclass
control to the respective tubes and place the antisera standardized for use with RBCs that we
tubes in the refrigerator. know of in the United States are those made by the
3. Wash the RBCs to be tested 4 times with cold Netherlands Red Cross. Their recommended
LISS using a refrigerated centrifuge (1000 × g for methods are a sedimentation or microplate (V
1 to 2 minutes). wells) AGT methods not generally used in the
4. Resuspend the RBCs to 3% to 5% in cold LISS. United States. Some years ago, we tested these anti-
5. Add 1 drop of RBCs to each of the two tubes sera for sensitivity and specificity using the routine
from the refrigerator. centrifugation AGT and a capillary tube test and
6. Mix and centrifuge in a serologic centrifuge for found the methods to be comparable.77 We found
the calibrated time. that some methods (e.g., albumin capillary method)
7. Inspect macroscopically for agglutination and that have been used to define the subclass of
record results. RBC-bound IgG were inappropriate and led to
8. Inspect macroscopically negative results, micro- false-positive results.77
scopically. Record results.
9. To the anti-IgG test, add 1 drop antiglobulin Sensitizing RBCs with Antibodies or Purified
control cells. Proteins of Known Subclass. Rh alloantibodies of
10. Centrifuge. known IgG allotype and subclass can be used to coat
11. Read macroscopically and record positive RBCs, or purified proteins of known subclass can be
results with a check mark. attached to chromic chloride-treated RBCs.

Quality Control Preparation of RBCs Coated with Known Subclass


The patient’s cold LISS washed RBCs should be 1. Using alloantibodies of known subclass: 0.2 mL
nonreactive with the diluent control. A positive test of appropriate dilutions of the antibodies are
with the control indicates the presence of a cold incubated at 37°C for 30 minutes with 0.1 mL
autoagglutinin and the test is invalid. of packed RBCs. The cells were then washed
Antiglobulin control cells, when added to the four times with saline and resuspended to a 5%
anti-IgG test, should be macroscopically reactive. If suspension.
not, then investigations need to include the quality 2. Using purified proteins of known subclass:
of the check cells, the efficiency of the washes, Protein quantification is performed by diluting
and/or the activity of the anti-IgG. Repeat the the purified proteins in 0.1 M NaOH and mea-
DAT. suring absorbance at a wavelength of 280 nm,
using a spectrophotometer. Protein concentra-
tions are calculated from a graph of known
Interpretation protein concentrations plotted against optical
1. A positive result with anti-IgG, in conjunction density at 280 nm. The samples are adjusted to
with a nonreactive diluent control, means that contain 1 mg/mL, and the procedure described
IgG is present on the patient’s RBCs. on pages 210 to 211 is followed.
2. When the routine DAT with anti-IgG is negative
or becomes weak/negative on sitting, and the Tube AGT Using IgG Subclass AGS
cold LISS wash DAT is positive or stronger, the Two drops of the IgG subclass antisera are added to
IgG is low affinity. 1 drop of a 5% washed sensitized RBC suspension,
214 Immune Hemolytic Anemias

mixed, and centrifuged at 1000 × g for 15 seconds. in vitro hemoglobinemia as being in vivo occur-
After centrifugation, the button of RBCs is gently rence. This will occur only in the clotted sample,
resuspended and inspected for agglutination (see as EDTA will inhibit any in vitro complement-
Table 6-2). mediated lysis.
4. If EDTA blood is not used, then in vitro comple-
Sedimentation AGT Using IgG Subclass AGS ment autosensitization may occur due to the pres-
One drop of IgG subclass antisera is added to 1 drop ence of normal incomplete cold autoantibodies.
of a 5% washed sensitized RBC suspension, mixed, This may lead to a false-positive result or an
and incubated at 37°C for 1 hour. After incubation, increase in the strength of the DAT. EDTA will
the tubes are gently removed from the water bath prevent any in vitro complement sensitization,
and the buttons inspected for a sedimentation even if the blood is cooled; thus, any complement
pattern. A smooth round button is considered a detected on the EDTA RBCs represents in vivo sen-
negative result, and a cell button with a broader sitization. Nevertheless, we do not routinely bleed
irregular border is considered a positive result. our patients into warmed syringes, etc., unless we
know we are dealing with a severe case of CAS or
Capillary AGT Using IgG Subclass AGS PCH. We find that by always warming the samples
Glass capillary tubes measuring 0.4 × 90 mm are before separation, we can obtain reliable results in
used. Equal volumes of IgG subclass antisera and most cases.
50% washed sensitized RBCs are drawn into the cap-
illary tube. The tubes are inverted and the end of the Determining the Blood Group
capillary containing antisera is inserted in a Sealease of DAT-Positive Patients
holder at a 45o angle. After 15 minutes at room tem-
perature, examine tubes for agglutination (beading, The main problem when typing DAT-positive RBCs is
ribbon, or granular appearance). Invert and examine that the RBCs may spontaneously agglutinate in the
again after 15 minutes. Repeat one more time. presence of potentiators, including albumin.78 In the
United States, manufacturers are allowed to add up to
8% albumin to any antisera; thus, we use a 10%
albumin negative control if a diluent control is not
GENERAL SEROLOGIC INVESTIGATIONS provided by the company and if the protein concen-
tration is not known. If monoclonal reagents are used,
The results obtained in each type of AIHA are dis- the protein concentration is usually lower (e.g., 3%)
cussed in Chapter 5 under the appropriate headings. and the negative control can be adjusted appropri-
ately. There should be no problems in ABO and Rh
Collection of Blood typing patients with AIHA if certain rules are fol-
lowed; however, if they are not followed, serious
We collect clotted blood to provide serum, and blood errors can occur. If cold autoantibodies are suspected,
in EDTA to provide RBCs for the DAT, ABO, and Rh the blood samples should be separated at 37°C, and
phenotyping and for preparation of eluates. If blood the RBCs should be washed with 37°C warm saline;
has not been kept at 37°C from the time of veni- if the patient has CAS, a 37°C centrifuge will have to
puncture, we incubate blood samples at 37°C for 10 to be used (see later).
15 minutes before we separate the serum (which is Garratty and coworkers78 studied how often spon-
separated by centrifugation at 37°C). We perform the taneous agglutination occurred with various media
DAT and prepare an eluate from the EDTA sample. when the RBCs had a positive DAT. Red cells from
Several problems can arise if the blood is not kept at 475 individuals (donors and patients) with a positive
37°C from the time of bleeding and cold autoanti- DAT (66% were 1⁄2 to 1+, 19% were 11⁄2+ to 2+, and
bodies are present: 15% were 21⁄2+ or greater) were examined for sponta-
neous agglutination after incubation in various
1. Autoagglutination of the RBCs may occur, leading media. Twenty-three percent of the samples showed
to difficulties in grouping. This almost always is a spontaneous agglutination in commercial Rh control
reversible phenomenon and can be overcome by solutions for “slide and rapid tube” reagents, 6% in
incubating the samples at 37°C before processing 30% albumin, 3% in 10% albumin, 2% in 6% albumin,
and testing. However, in rare cases, autoagglutinin and 1% in saline. Cells suspended in serum were
may persist at 37°C. more prone to spontaneous agglutination. The
2. Loss of antibody from the serum by in vitro autoad- strength of spontaneous agglutination varied; more
sorption, leading to a false low cold agglutination than 50% of the samples reacted only 1+ or less,
titer, etc. This is also a reversible phenomenon (see approximately 10% reacted greater than 2+. There
later). was not a complete correlation with spontaneous
3. Possible direct hemolysis of the RBCs, using anti- agglutination and the quantity of IgG on the cells, as
body and complement, again leading to a false low determined by the AGT. Of the samples showing
laboratory value and possible misinterpretation of spontaneous agglutination, 50% were associated
The Serologic Investigation of Autoimmune Hemolytic Anemia 215

with a DAT strength of more than 2+, 27% with a agglutinins will usually react well at 37°C but the cold
DAT of 11⁄2+ to 2+, and 23% with a DAT of 1⁄2+ to 1+. autoantibody will not react. These results may be
Spontaneous agglutination occurred with the same confirmed (by the normal methods) after the cold
frequency whether the cells were sensitized with autoantibody has been autoadsorbed from the
both IgG and C3 or with IgG alone. Surprisingly, 7% patient’s serum. As patients with WAIHA do not often
of the samples sensitized with C3 but no detectable have strong autoagglutinins, problems in ABO typing
IgG also demonstrated spontaneous agglutination; are rare.
this may have been due to RBC-bound IgM. Marked Rh Phenotype. It is useful to Rh phenotype patients
differences in reaction strength were seen with Rh with WAIHA, preferably before they are transfused.
control solutions from different manufacturers, and Monoclonal antisera are preferable to polyclonal
the degree of spontaneous agglutination was incon- reagents as they contain less protein or potentiators,
sistent with individual products.78 so fewer false-positive results due to spontaneous
Most monoclonal reagents do not usually contain agglutination occur. Commercial “slide and rapid
potentiators and usually give reliable results, but tube” reagents always contain potentiators (e.g.,
problems due to potentiators have been reported.79 albumin) and the manufacturers should always make
Because of this, and because spontaneous agglutina- available the diluent containing the potentiator but
tion can occur in low protein solutions (see earlier),78 not the anti-Rh; this must always be tested in parallel
we prefer to have a negative control. Ideally, this as a negative control. If the diluent control is negative
should be the diluent without the Rh antibody; if this then the Rh typing result on the patient is acceptable,
is not available then a negative control containing even if the DAT is strongly positive. If the control is
albumin equivalent to the protein concentration of positive, then the Rh typing will have to be repeated
the antiserum should be used. If this information is with a “saline tube test” anti-Rh or monoclonal
not available, then we recommend using 8% to 10% reagents. These reagents are said not to give false-pos-
albumin as a control. Rodberg and coworkers79 itive reactions with RBCs, even strongly sensitized
reported that one company’s monoclonal Rh antisera with IgG. Nevertheless, we believe false-
gave problems with DAT+ patients because a poten- positive reactions may still occasionally occur and we
tiator was present. Monoclonal anti-Rh reagents were prefer to always use an albumin negative control
used according to manufacturer’s directions on when using such reagents.
untreated and chloroquine-treated DAT+ RBCs. Of
32 DAT+ patients, who appeared to be D+C+c+E+e+
(R1R2), 13 still appeared to be R1R2 following chloro- Phenotyping DAT+ RBCs When
quine treatment; but 19 appeared to be R1R1, R2R2, Spontaneous Agglutination Occurs
Ror, or rr following testing of chloroquine-treated or When Using Antiglobulin Reactive
RBCs. None of these DAT+ RBCs reacted in the 6% Antisera
albumin control. RBCs from 2 other DAT+ (3+)
patients reacted weaker (1+) than expected with Spontaneous agglutination usually is associated with
monoclonal anti-e and reacted stronger (3+) after IgG sensitization but on rare occasions is associated
chloroquine treatement (DAT still 2+). These latter with RBC-bound IgM (see Chapter 9).
results suggest that blocking auto anti-e might have Most of the phenotyping approaches are based on
caused the initial weakly positive result with anti-e. methods used to remove IgG from RBCs, but IgM
Thus, fragile, or weaker than expected, agglutination may also be removed. The method should cause
results obtained when Rh typing DAT+ RBCs with minimal or no damage to RBC antigens; careful con-
monoclonal anti-Rh reagents must be interpreted trols for this are essential. The first method used was
with care. Such results might indicate a false positive, gentle heat (e.g., 45°C to 50°C) for 5 to 10 minutes.
or a true weak positive due to a blocking auto- This is not a very efficient method compared with the
antibody. The false-positive results were not indi- later methods but can be used if careful controls are
cated by the 6% albumin control recommended by used.
this particular manufacturer. We prefer the EDTA-glycine acid method, which
ABO Group. The washed RBCs are tested in the relies on low pH. The method, described by Louie and
usual fashion with anti-A, anti-B, and perhaps coworkers,80 is based on an elution method described
anti-A,B, but a negative albumin control is also set up. by Rekvig and Hannestad81 and is the basis of a com-
This control should be compared with the tests and if mercial kit called “EGA” (Gamma Biologicals). The
positive indicates either nondispersed autoagglutina- method is efficient for typing Rh and most other
tion or spontaneous agglutination of heavily IgG- antigens, but it should be noted that Kell antigens are
sensitized cells in albumin. destroyed or greatly diminished. des Roziers and
The patient’s serum is tested against A1, B, O, and Squalli82 found the method of Louie and coworkers80 to
his own cells as usual. If the patient is known to have be superior to heat and chloroquine methods. A
or appears to have CAS, the serum typing tests should modification of the Louie method80 by Byrne83 uses
be repeated by a prewarmed technique, strictly at larger volumes of reagent so that an eluate suitable for
37°C. At this temperature, all but the weakest ABO testing is also obtained in addition to the treated RBCs.
216 Immune Hemolytic Anemias

Chloroquine has been used to inhibit RBC antigen- 7. Using 1.0 M Tris and pH paper, carefully adjust
antibody reactions in vitro and in vivo.84,85 Mantel the pH of the eluate to between 7.0 and 7.4.
and Holtz85 used chloroquine to elute IgG autoanti- (Caution: 1.0 M Tris-NaCl is very alkaline, and
bodies from RBCs of patients with AIHA. Edwards only a very small amount is required to attain the
and coworkers86 applied this to typing RBCs with a desired pH.)
positive DAT. Complete dissociation of IgG antibody 8. If a precipitate forms in the eluate, centrifuge the
occurred in 47 of 56 strongly sensitized RBC samples eluate and remove the clear supernatant (eluate)
from patients with positive DAT. They did not into another labeled tube.
observe any apparent loss of ABH, Rh, MNSs, P1, 9. The eluate may now be used for testing in paral-
Lewis, Kell, Duffy, or Kidd antigens. Other workers,87- lel with the last wash.
89 including ourselves, have observed weakening of 10. The acid/EDTA-treated RBCs should be washed
some blood group antigens after chloroquine treat- at least 3 times in 0.9% sodium chloride before
ment. Fortunately, this is not sufficient to be noticed use, and tested by the DAT. If the DAT is nega-
when using strongly reactive antisera, but if weaker tive, the RBCs are ready for use. If the DAT is
antisera are used, then the reactions may be much still positive, repeat treatment (steps 1 to 6) one
weaker than with untreated RBCs. This does not make more time (for a maximum of 2 treatments).
the technique any less useful as long as weak positive
control RBCs (e.g., from heterozygotes) are incubated
along with the test cells undergoing the chloroquine
treatment. DISSOCIATION OF IgG AUTOANTIBODY
USING CHLOROQUINE REAGENT86

DISSOCIATION OF IgG AUTOANTIBODY Materials


USING A MODIFIED GLYCINE-ACID/EDTA PBS, pH 7.3
TECHNIQUE80 Chloroquine diphosphate, 200 mg/mL
20 g chloroquine diphosphate in 100 mL PBS
Adjust pH to 5.0 ± 0.1 with 1N or 5N NaOH
Reagents Store at 2°C to 8°C for no more than 1 year
10% EDTA [disodium ethylenediamine-tetraacetate Anti-IgG
(Na2EDTA)] Positive and negative control RBCs for the antigen(s)
0.1 M glycine acid buffer (pH 1.5) to be tested
0.75 g glycine Test tubes
Make up to 100 mL with 0.9% sodium chloride Serologic centrifuge
Adjust to pH 1.5 using concentrated hydrochloric Antiglobulin control cells
acid (HCl)
1.0 M Tris-NaCl Procedure
12.1 g Tris(hydroxymethyl)aminomethane (Tris) 1. Wash the test and control RBCs in PBS
5.25 g sodium chloride 2. To 1 vol of washed, packed RBCs, add 4 vol of
Make up to 100 mL with distilled water chloroquine.
3. Mix and incubate at room temperature for
Procedure 30 minutes.
1. Wash RBCs 6 times in 0.9% sodium chloride. On 4. Remove a small amount of the chloroquine-
the last wash, pack the RBCs well and transfer treated test RBCs and wash 4 times in PBS.
all supernatant to a tube labeled “last wash.” 5. Test with anti-IgG.
2. In a test tube, mix together 20 vol of 0.1 M If the anti-IgG is nonreactive, wash the test and
glycine-HCl buffer (pH 1.5) and 5 vol 10% control RBCs 4 times in PBS and use for pheno-
EDTA. This is the elution reagent. typing.
3. Place 10 vol of packed RBCs that have been If the anti-IgG is reactive, repeat steps 4 and
washed 6 times in 0.9% sodium chloride into a 5 at 30-minute intervals until the anti-IgG is non-
12 × 75-mm glass tube. reactive or up to 2 hours.
4. Add 20 vol of elution reagent to the RBCs, mix If the anti-IgG is still reactive after 2 hours of
well, and incubate at room temperature for treatment, use a different approach to remove
2 minutes. (Caution: Overincubation will cause the IgG (e.g., glycine acid/EDTA).
irreversible damage to the RBCs.) 6. Add antiglobulin control cells to all negative
5. After incubation add 1 vol of 1.0 M Tris-NaCl, tests. Centrifuge and examine for agglutination.
mix, and immediately centrifuge at 1000 × g for
60 seconds. Quality Control
6. Remove supernatant (now the eluate) into a Control RBCs must be treated in the same way as the
labeled tube. test RBCs and must give valid typing results. If the
The Serologic Investigation of Autoimmune Hemolytic Anemia 217

antigen activity of the positive control RBCs is SEROLOGIC INVESTIGATIONS


diminished, use a different approach to remove IgG TO HELP IN THE DIFFERENTIAL
from the test RBCs (e.g., glycine acid/EDTA). DIAGNOSIS OF AIHA
If the antiglobulin control cells are not macrosco-
pically positive or react weaker than expected, then
repeat the test. In this section, we describe detailed screening tests
that may be used in evaluating a patient with pos-
Interpretation sible AIHA. All tests are not necessary in all cases,
When the procedure is effective, IgG is completely although care must be taken in the interpretation of
removed from the IgG-coated RBCs and pheno- abbreviated serologic evaluations. The significance of
typing is possible. each step of the diagnostic workup is further dis-
cussed in Chapter 5.
Notes To diagnose and classify the patient correctly,
1. Incubation of RBCs with chloroquine diphos- several questions have to be answered:
phate should not exceed 2 hours; hemolysis or QUESTION 1: Are the patient’s RBCs sensitized with
loss of antigen activity could occur. protein?
2. IgM is removed, but C3 is not removed from the To answer this question, the patient’s washed RBCs
RBCs by chloroquine. are first tested with a polyspecific AHG. Most com-
3. When testing Rh antigens, use high-protein mercial AHG will detect IgG sensitization adequately,
reagents. It has been reported that Rh antigens but approximately 20% to 30% of all AIHA cases have
can be weakened by chloroquine treatment, only complement (C3d and C4d) on their RBCs, and
especially when saline or chemically modified rare cases have been described with only IgA or IgM
Rh reagents are used. on their cells; as mentioned previously, some commer-
4. This method is also effective in removing Bg cial polyspecific AGS will not detect IgA and IgM and
(HLA-related) antigens from RBCs. occasionally will not detect weak C3d sensitization.
Other cases exist (perhaps as many as 5% to 10% of
all AIHA cases), where a negative DAT is found in the
PHENOTYPING DAT-POSITIVE RBCS presence of AIHA (see Chapter 9). Thus, if the DAT is
BY ADSORPTION OF ANTISERA negative and the patient has all the symptoms of
AIHA, the diagnosis of AIHA should not be excluded
on the results of the DAT.
Sometimes the IgG autoantibody will not dissociate
after treatment. If this happens, typing can be carried
out by measuring the amount of specific antibody DAT
left in the typing antiserum after adsorption with the
patient’s RBCs, and comparing the results with
adsorptions using RBCs known to contain the appro- 1. Wash RBCs (preferably from EDTA blood) 4 times
priate antigens (RBCs from heterozygotes and in large volume of saline. (We usually wash 2 or
homozygotes). 3 drops of whole blood.)
2. Add 1 drop of 2% to 5% washed RBCs to 2 drops
Procedure of polyspecific AHG (or add 2 drops of AHG to
1. Add 0.2 mL of the antiserum to 0.2 mL washed, the dry button from 1 drop of 2% to 5% washed
packed RBCs from the patient. To other 0.2-mL RBCs).
aliquots of the antiserum, add 0.2 mL antigen- 3. Centrifuge tubes at 1000 × g RCF for 15 to 20 sec-
negative and antigen-positive RBCs, respec- onds (time specified by serologic calibration of
tively. Mix. centrifuge).
2. Incubate the three adsorption tests at 37°C for 4. Inspect tubes for agglutination (we prefer to
1 hour. Mix occasionally. check all macroscopically negative results for
3. Centrifuge the three tubes and separate the microscopic agglutination).
serum into labeled tubes.
4. Label three sets of 10 × 75-mm tubes. Make Controls
twofold dilutions (0.1 mL) of each aliquot of We always add 1 drop of the washed patient’s RBCs
adsorbed antiserum using saline as a diluent. to 2 drops of the AHG diluent (or 5% to 10% albumin
5. To each dilution of adsorbed antiserum, add if diluent is not available), as a negative control, and
0.1 mL 2% to 5% saline-suspended antigen- centrifuge this along with the test. The DAT is only
positive RBCs. truly positive if the AHG reacts stronger than the
6. Incubate the 3 sets of titration tubes for 30 minutes diluent; ideally, of course, the tube containing
at 37°C. Perform AGT on RBCs from all tubes. diluent should be negative. If the negative control is
7. Compare the results (Table 6-3). positive, it could be due to cold autoagglutinins or
RBC-bound warm IgM autoantibody. To dispense
218 Immune Hemolytic Anemias

TABLE 6-3. Jka TYPING OF PATIENT WITH POSITIVE DIRECT ANTIGLOBULIN TEST
USING DIFFERENTIAL ADSORPTION

Dilutions of Adsorbed Anti-Jka Tested Against


RBCs Used to Adsorb Jk (a+b+) RBCs*
Anti-Jka Typing Serum 1 2 4 8 16 32 Score

Jk(a+b+) 2+ 1+ 1+ 1⁄2+ 0 0 17
Jk(a+b–) 1+ 1+ 0 0 0 0 8
Jk(a–b+) 3+ 3+ 2+ 2+ 1+ 0 32
Patient’s 2+ 1+ 0 0 0 0 10

* These results indicate that the patient is probably Jk(a+)

cold autoagglutinins, the EDTA sample should be QUESTION 3: Does the serum contain antibodies?
warmed to 37°C and the RBCs washed with 37°C Are they agglutinins or “incomplete”? Do they have
saline. If the control is still positive, this suggests the hemolytic activity? At what temperature do they react
presence of an IgM warm autoantibody and the optimally? What is their thermal range? What is their
RBCs may have to be treated with 2-mercap- specificity? Are they autoantibodies or alloantibodies?
toethanol (2ME)90 or dithiothreitol (DTT) to prevent These questions can be answered by regular sero-
spontaneous agglutination. logic techniques (with a few minor additions) used in
Red cells presensitized with IgG (e.g., anti-D) the blood bank for detection and identification of
should always be added to all negative tests with antibodies.
AHG. After recentrifugation, the tests should now be To aid in the visualization of hemolysis, we use a
positive, ensuring that the AGS has not been inhi- 10% RBC suspension; to keep the antigen-antibody
bited (e.g., by inadequate washing). No AGT should ratio in a ratio similar to usual, we add 1 vol of the cell
ever be called negative without this control. suspension to 4 vol of serum. As patients with AIHA
may have low serum complement levels, it is advis-
able to set up a duplicate set of tests to which an equal
QUESTION 2: What proteins are present on the RBCs? volume of fresh compatible inert serum has been
The RBCs are often sensitized with IgG and com- added as a source of complement. The mixture of
plement or IgG alone, and sometimes by complement patient’s serum and complement should be in the pH
alone. The presence of these proteins is best detected range 6.5 to 6.8 (i.e., add 0.1 vol of 0.2N HCl to the
by performing AGTs using monospecific AGS such as serum), as this seems to be optimal for the detection of
anti-IgG and anticomplement containing anti-C3d. warm and cold hemolysins.
Red cell–bound IgA and IgM are sometimes detected Many warm and cold reacting autoantibodies give
but they are usually present together with IgG and/or enhanced reactions with enzyme-treated RBCs; thus,
complement. it is useful to include such cells in the serum screening
The amount of protein sensitizing the RBCs can be procedure. Hemolysis of enzyme-treated cells is much
judged in a semiquantitative way. By testing the RBCs more commonly observed than hemolysis of
of the patient with dilutions of the AHG, one can untreated RBCs.
obtain a relative estimate of the amount of protein It is extremely important that tests set up at 37°C are
sensitizing the RBCs. For instance, the undiluted AHG kept strictly at 37°C. To do this, the patient’s serum,
may show 3+ results on two specimens, but the two reagent RBCs, and any other reagents (e.g., albumin)
may have very different agglutination scores on titra- are warmed to 37°C separately before mixing. The
tion. Agglutination scores correlated very well with tests are then centrifuged at 37°C, and the cells are
results of assays that measured the number of washed in 37°C saline for the AGT. If this is not done
RBC-bound IgG and C3 molecules. Although we have carefully (with careful control of the temperature at
previously emphasized that commercial AGS should each stage), then positive results may be misinter-
never be diluted when used routinely (e.g., for com- preted, lending to erroneous conclusions regarding
patibility testing), it is acceptable to dilute these anti- the classification of the AIHA and to the clinical
sera when they are only being used to compare the significance of the autoantibody.
amount of RBC-bound IgG or C3. We stress once The approach we give below is optimal; we have
again, they must never be diluted when used for anti- been rewarded many times by the extra work
body detection or crossmatching. More accurate ways involved. On the other hand, a great deal of informa-
of measuring the amount of RBC-bound proteins tion can be obtained by routine blood bank techniques
using radiolabeled AHG, ELISA, and flow cytometry (e.g., a DAT using anti-IgG and anti-C3d; antibody
have been described but are rarely useful at a practi- screening with one set at RT and one set at 37°C; a
cal clinical level.91-94 cold agglutinin titer at 4°C if RT agglutination is
The Serologic Investigation of Autoimmune Hemolytic Anemia 219

obtained and one tube test of patient’s serum plus Procedure


RBCs plus 30% albumin at 30°C). 1. Turn on 37°C centrifuge to warm up. Place 37°C
The serum may contain: PBS into a 37°C water bath to warm up. Turn on
pH meter.
• No antibodies; they may all have been autoadsorbed 2. Label two tubes for pooled RBCs, “PC” or
onto the patient’s RBCs in vivo “pool,” and another 2 tubes for autologous cells,
• Only autoantibodies “Auto.” Label one of each pair: “UT” (untreated)
• Autoantibodies plus alloantibodies or “ET” (enzyme-treated).
• Alloantibodies only 3. Place 16 drops of 3% to 4% pooled RBCs into
each of the two tubes marked “PC” or “pool.”
4. Place 16 drops of 3% to 4% washed patient’s
RBCs into each of the two tubes marked “Auto.”
SERUM SCREEN TO DETERMINE SERUM
5. Wash RBCs 1 time with PBS.
ANTIBODY(IES) CHARACTERISTICS
6. Add 2 vol of 0.1% ficin to 1 vol of the packed
RBCs in the “ET” tubes (e.g., 0.2 mL ficin added
Purpose to 0.1 mL RBCs).
To characterize serum autoantibodies in AIHA as far 7. Incubate at 37°C for the specified time for that
as the following: Are they agglutinins or sensitizing lot (e.g., 15 minutes).
antibodies? Do they have hemolytic activity? Do 8. Wash 3 times with PBS.
they react preferentially at 20°C or at 37°C? Results 9. Add 6 drops of PBS to each of the packed RBCs
of this testing, in conjunction with results of the DAT in the “UT” tube and the “ET” tube.
and the titer/thermal amplitude, can aid in the dif- 10. Place 27 drops of complement into a 12 × 75-mm
ferential diagnosis of WAIHA versus cold AIHA tube labeled “AC.” Add 3 drops 0.2N HCl.
versus warm plus cold AIHA variations. 11. Place 9 drops of patient’s serum into a 12 ×
75-mm tube labeled “ASAC.” Add 1 drop 0.2N
Principle HCl and 10 drops of AC from step 10.
The patient’s serum is tested with and without 12. Place approximately 1 mL patient’s serum into a
acidification and the addition of fresh normal serum 12 × 75-mm tube labeled “S.”
(as a source of complement), against untreated and 13. Determine pH of sera in “S,” “ASAC,” and “AC”
enzyme-treated RBCs (allogeneic and autologous), tubes. Record results on the worksheet.
at 20°C and 37°C (prewarmed). 14. If the pH of the sera in the “AC” or “ASAC”
tubes is less than 6.5 or greater than 6.8, then
Specimen Requirements adjust accordingly by adding more comple-
Patient’s serum, about 2 mL ment, or complement plus patient’s serum
Patient’s RBCs, preferably in EDTA; 37°C washed, or (± acidification), respectively. In adjusting the
DTT/2ME-treated, if appropriate. pH of the ASAC, it is important to keep the 1:1
ratio of complement to serum. Record pH and
Materials any adjustment made.
10 × and 12 × 75-mm test tubes 15. Label two sets of eight 10 × 75-mm tubes: two
Complement source [pooled normal donor sera, not tubes are labeled “S” (patient’s serum), two
containing unexpected antibodies, fresh or stored (in tubes are labeled “ASAC” (acidified patient’s
aliquots) at –70°C. serum plus AC), two tubes are labeled “AC”
RBCs [usually a pool of two commercial antibody (acidified complement), and two tubes are
detection RBCs (e.g., R1R1 + R2R2)]. If patient has labeled “Auto” (autologous RBCs). One of each
alloantibodies, then RBCs should lack antigens that pair is labeled “UT” (untreated cells), and the
would react with alloantibody. other is labeled “ET” (enzyme-treated cells). One
0.1% Ficin, prepared fresh by adding 4.5 mL PBS to set of eight tubes is labeled “20°C,” and the other
0.5 mL of 1% ficin set of eight tubes is labeled “37°C.”
37°C water bath 16. Place 4 drops of patient’s serum into each of the
20°C incubator if available four tubes labeled “S” and into each of the four
0.2N HCl: 10 mL 1N HCl + 40 mL deionized H2O tubes labeled “Auto.”
pH meter with pH 7 reference buffer 17. Place 4 drops of acidified serum plus AC (ASAC)
PBS, pH 7.3 into each of the four tubes labeled “ASAC.”
Uniform drop glass pipettes 18. Place 4 drops of the AC into each of the four
Serologic centrifuge tubes labeled “AC.”
37°C serologic centrifuge (if available) 19. Place the set of serum tubes labeled “37°C”
37°C PBS in squeeze bottle into the 37°C water bath. Place the set of serum
Polyspecific AGS tubes labeled “20°C” and the RBC tubes at 19°C
Antiglobulin control cells to 21°C.
220 Immune Hemolytic Anemias

20. After the 20°C sera tubes and the RBC tubes 40. Add antiglobulin control cells to all negative
have reached 19°C to 21°C (e.g., ≥5 minutes’ tests.
incubation, if necessary), use a uniform drop 41. Centrifuge.
pipette to add 1 drop of untreated pooled RBCs 42. Read for agglutination (macroscopically) and
to each of the S, ASAC, and AC tubes labeled record results.
“UT”; 1 drop of the untreated autologous RBCs
to the tube labeled “Auto UT”; 1 drop of the Quality Control
enzyme-treated pooled RBCs to each of the S, Antiglobulin control cells are added to check for
ASAC, and AC tubes labeled “ET”; and 1 drop of reactivity/presence of AGS. Reactivity should be
the enzyme-treated autologous RBCs to the tube macroscopically positive. If the antiglobulin control
labeled “Auto ET.” Record temperature on cells are nonreactive or react weaker than expected,
worksheet. then repeat the test.
21. Move the RBC tubes to 37°C. Make sure a The AC tests control for reactivity of the comple-
uniform drop pipette is in each tube. ment source in the test system and are usually non-
22. After the 37°C sera tubes and the RBC tubes reactive. If the AC does react, then results of patient’s
have reached 37°C (≥5 minutes’ incubation), add serum plus AC need to be judged taking that back-
RBCs to sera tubes as in step 20. Record temper- ground reactivity into account.
ature on worksheet.
23. Incubate all tubes for 1 to 2 hours. Interpretation
24. Gently resuspend the “20°C” set of settled RBCs Results are interpreted on a case-by-case basis, in
and record the results (settled reading). conjunction with the results of the titer/thermal
25. Centrifuge the “20°C” set for 15 to 20 seconds amplitude study (if performed) and the DAT. Typical
(previously calibrated time) at 1000 × g. results for warm AIHA and CAS are shown in Tables
26. Observe the supernatant carefully for hemolysis, 6-4, 6-5, and 6-6.
and record results.
27. Gently resuspend the button of cells and Notes
observe for agglutination. Record results after 1. If the patient’s serum contains alloantibodies,
centrifugation. select RBCs that are antigen negative. If the
28. Gently resuspend the “37°C” set of settled RBCs patient’s autoantibody shows a blood group
and record the results (settled reading). (Read specificity, then select antigen positive test
each tube one at a time, returning tubes to 37°C RBCs.
as soon as possible.) 2. If there is no reactivity observed in the 20°C
29. Bring the 37°C centrifuge to 37°C ± 2°C by tests, then there is no need to spin or wash the
centrifuging (e.g., 1 minute or more). “37°C” tests at 37°C. A cell washer could be used
30. Centrifuge the “37°C” set for 15 to 20 seconds to wash RBCs for the AGT, however, still remove
(calibrated time) in the 37°C centrifuge. the serum before washing and reduce the RBC
31. Maintain the 37°C temperature (i.e., by keeping volume after washing as in steps 34–37.
the tubes in the warm 37°C centrifuge or placing Performing the IAT on the 10% RBC button can
them back into the 37°C incubator). lead to decreased sensitivity.
32. Carefully observe each supernatant for hemo- 3. If the 37°C tests are not performed strictly at
lysis. Record results. 37°C, then positive results may be misinter-
33. One tube at a time, gently resuspend the button preted, leading to erroneous conclusions regard-
of cells and observe for agglutination (return ing the classification of the AIHA and to the
tubes to the 37°C incubator or 37°C centrifuge as clinical significance of the autoantibody.
soon as possible). Record results after centrifu-
gation.
34. Recentrifuge the “37°C” set for 30 to 60 seconds
in the 37°C centrifuge. COLD AGGLUTININ TITER/THERMAL
35. Quickly remove supernatant serum. AMPLITUDE/Ii SPECIFICITY
36. Wash RBCs 4 times with 37°C saline in the 37°C
centrifuge.
37. Add 2 drops PBS to each tube. Draw RBCs up We usually only perform these tests if agglutination
into pipette and dispense 1 drop back into the is observed at 20°C in the serum screening. If only
tube; discard the pipette containing the remain- the cold agglutinin titer is required, the tests below
der of the RBCs. can be reduced to a single row of dilutions tested
38. Add polyspecific AGS to each of the tubes. against group OI adult RBCs incubated at 4°C. Pre-
39. Centrifuge for 15 to 20 seconds (calibrated time) cautions mentioned below in step k. still apply.
at 1000 × g. Read for agglutination (macroscopi- 1. Serial dilutions (1 in 1 to 1 in 2048) of the warm
cally negative tests should be read microscopi- (37°C) separated patient’s serum are prepared in
cally). Record results. saline, in 0.5 mL quantitities. The last aliquot to be
The Serologic Investigation of Autoimmune Hemolytic Anemia 221

TABLE 6-4. TYPICAL RESULTS OF SERUM SCREENING TESTS IN AUTOIMMUNE


HEMOLYTIC ANEMIA

Warm Autoimmune Hemolytic Anemia

S AS + AC Negative Control AC Auto

Untreated RBCs 20°C Lysis 0 0 0 0


Agglutination 0 0 0 0
37°C Lysis 0 0 0 0
Agglutination 0 0 0 0
IAT 2+ 2+ 0 *
Enzyme-treated RBCs 20°C Lysis 0 0 0 0
Agglutination 1+ 1+ 0 1+
37°C Lysis 1⁄2+ 2+ 0 1⁄2+
Agglutination 3+ 3+ 0 3+

S, Patient’s serum; AS, patient’s serum + 0.1 vol 0.2 N HCl, pH 6.5 to 6.8; AS + AC, Acidified serum + acidified complement (normal
inert fresh serum + 0.1 vol 0.2 N HCl, pH 6.5 to 6.8.
*Depends on strength of DAT.

TABLE 6-5. TYPICAL RESULTS OF SERUM SCREENING TESTS IN AUTOIMMUNE


HEMOLYTIC ANEMIA

Cold Agglutinin Syndrome

S AS + AC Negative Control AC Auto

Untreated RBCs 20°C Lysis 1+ 3+ 0 0


Agglutination 4+ 4+ 0 4+
37°C Lysis 0 0 0 0
Agglutination 0 0 0 0
IAT 0 0 0 *
Enzyme-treated RBCs 20°C Lysis 2+ 4+ 0 1+
Agglutination 4+ X 0 4+
37°C Lysis 0 0 0 0
Agglutination 0 0 0 0

S, Patient’s serum; AS, patient’s serum + 0.1 vol 0.2 N HCl, pH 6.5 to 6.8; AS + AC, acidified serum + acidified complement (normal
inert fresh serum + 0.1 vol 0.2 N HCl, pH 6.5 to 6.8; X, no RBCs left.
*Depends on strength of DAT.

TABLE 6-6. TYPICAL RESULTS OF SERUM SCREENING TESTS IN AUTOIMMUNE


HEMOLYTIC ANEMIA

Paroxysmal Cold Hemoglobinuria

S AS + AC Negative Control AC Auto

Untreated RBCs 20°C Lysis 0 0 0 0


Agglutination 0 0 0 0
37°C Lysis 0 0 0 0
Agglutination 0 0 0 0
IAT 0 0 0 ‡
Enzyme-treated RBCs 20°C Lysis 0 2+† 0 0
Agglutination 1+* 1+* 0 0
37°C Lysis 0 0 0 0
Agglutination 0 0 0 0

S, Patient’s serum; AS, patient’s serum + 0.1 vol 0.2 N HCl, pH 6.5 to 6.8; AS + AC, acidified serum + acidified complement (normal inert
fresh serum + 0.1 vol 0.2 N HCl, pH 6.5 to 6.8).
*Found in 3 of 6 cases we studied.
†Found in 4 of 6 cases we studied.
‡Depends on strength of DAT.
222 Immune Hemolytic Anemias

TABLE 6-7. EXAMPLE OF RESULTS OF A COLD AGGLUTININ TITER

Dilutions of Patient’s Serum (×103)

1 2 4 8 16 32 64 128 256

Same pipette or pipette tip 4+ 4+ 4+ 4+ 3+ 3+ 2+ 1+ 0


Separate pipettes or pipette tip 31⁄2+ 3+ 2+ 1+ 1⁄2+ 0 0 0 0

carried over is kept in case further dilutions are refrigerated centrifuge, validated for serologic
required. It is of prime importance that separate methods), after 30 minutes to 1 hour, or left for at
pipettes (or pipette tips) are used to transfer each least 2 hours (we often leave ours overnight) to
0.5 mL aliquot to the next tube when preparing sediment at 4°C before reading.
each dilution as “carry over” can give extremely When reading the 4°C tests, it is important
inaccurate high titers if a single pipette or pipette that the rack of tubes is placed in a melting ice
tip is used. (See Table 6-7; patient’s correct titer bath (following centrifugation or sedimenta-
was 8000, not 128,000 as reported to us.) tion) and the tubes read rapidly, one at a time.
2. Place three rows of 10 × 75 mm tubes in a rack. Table 6-7 shows results from a typical anti-I cold
The first row is labeled “I.” The second row is autoagglutinin.
labeled “i” and the third row is labeled “auto.”
3. Starting from the highest dilution (e.g., 2048),
Interpretation
dispense 2 drops of diluted serum into three
1. Normal cold agglutinin titer is less than 64 in
tubes (e.g., 2048I; 2048i and 2048 auto).
saline at 4°C.
4. Prepare three 2% to 5% washed RBC suspensions
2. Cold agglutinin titers associated with CAS are
from group O adult I cells (e.g., pool of two
usually ≥1000 at 4°C.
screening cells), group O cord i cells, and the
3. Reactivity of a cold agglutinin at 30°C in
patient’s own 37°C washed RBCs. In addition, if
albumin distinguishes clinically significant (i.e.,
the patient is group A or B, it is sometimes useful
associated with in vivo hemolysis) from clini-
to test A1 or B RBCs (this may suggest anti-H, -HI,
cally insignificant cold autoantibodies.95 If the
-A or -B specificity instead of the more common
patient has hemolytic anemia and the agglutinin
anti-I specificity).
was nonreactive at 30°C, perform the 30°C test in
5. Incubate the tubes containing serum dilutions at
the presence of albumin (i.e., patient’s undiluted
37°C until they reach 37°C (5–10 minutes).
serum + 30% albumin + RBCs95) to determine
6. Incubate aliquots of the washed RBC suspen-
the thermal amplitude.
sions at 37°C, along with the serum dilutions,
4. Cold agglutinins of normal titer at 4°C (≤64), but
and also allow to reach 37°C.
of high thermal amplitutde (30°C or 37°C) can be
7. When both RBCs and serum are at 37°C, add
seen in the “mixed type” AIHA (i.e., IgG warm
1 drop of cell suspension to each of the appro-
autoantibodies are also present).
priate rows. Mix and allow to incubate for up to
5. Some agglutinins in the IgM warm autoimmune
2 hours.
category demonstrate optimal reactivity at
8. Either centrifuge strictly at 37°C after 30 minutes
20°C to 30°C.
to 1 hour or allow the RBCs to sediment at 37°C
6. A titer difference of more than two tubes and/or
for 2 hours before reading for agglutination. If the
a score difference of greater than 10 is considered
tubes are centrifuged at 37°C, they must be put
to indicate a significant difference between
back in the 37°C water bath immediately after
results of different RBCs.
centrifugation and each tube read separately, as
7. Stronger reactions with I adult RBCs than with
quickly as possible, so that no cooling ensues.
cord RBCs and/or i adult RBCs indicate anti-I
Similar care must be taken if the cells are
specificity.
allowed to sediment.
8. Stronger reactions with cord RBCs than with
9. After reading, move the tubes to a 30°C water
I adult RBCs indicate anti-i specificity.
bath. Then read the tests following 2 hours
9. Anti-IT reacts strongest with i cord RBCs, less
sedimentation at 30°C, taking the same care as
strong with I adult RBCs, and least strong with
mentioned above.
i adult RBCs.
10. Next, incubation at room temperature is used;
then the tubes can be centrifuged at room tem-
perature after 30 minutes to 1 hour. Notes
11. After reading, incubate the tubes at 4°C. The 1. If only a cold agglutinin titer is required, testing
tubes can either be centrifuged at 4°C (i.e., in a can be limited to a single set of dilutions tested
The Serologic Investigation of Autoimmune Hemolytic Anemia 223

against a pool of group O adult I RBCs incubated DONATH-LANDSTEINER (DL) TEST


at 4°C for 1–2 hours or overnight.
2. Use separate pipette tips for transferring each
aliquot when preparing dilutions as carryover Procedure
can result in inaccurate titers. 1. Place 5 drops of patient’s serum into each of
3. It may be desirable to test other RBCs (e.g., i 4 tubes, two labeled “test” and two labeled
adult RBCs or ABO identical RBCs) in order to “control.”
determine the autoantibody specificity. 2. To one of the “test” tubes and one of the
4. Thoroughly mix the patient’s serum/plasma “control” tubes, add 2 to 5 drops of fresh normal
before preparing the dilutions. If cryoglobulins serum.
are present and precipitated out, the titer result 3. To a 5th tube (complement control), add 5 drops
may not be accurate (falsely lower). of fresh normal serum.
5. Settled readings are more accurate after 2 hours 4. Add 1 drop of 25% RBCs to each tube.
of incubation. 5. Place the patient’s “control” tubes in the 37°C
6. If the agglutinin reacts at the warmer tempera- incubator.
tures (e.g., at 37°C or 30°C) and the titer is not 6. Place the patient’s “test” tubes and the comple-
increasing as the temperature is lowered, then ment control tube in a melting ice bath for
the observed titers at the lower temperatures 30–60 minutes.
may be due to agglutination carryover. It may be 7. After 30–60 minutes, gently mix the tubes in the
necessary to repeat the titers with separate sets melting ice and transfer them to the 37°C incu-
of tubes for each temperature. bator. Incubate 30 minutes.
8. Gently mix and centrifuge all tubes.
Titration of Hemolysins 9. Observe for lysis. Record results (hemolysis is
If hemolysins are detected in the serum screen, it is graded on a 0 to 4+ scale with 4+ indicating total
not usually necessary to quantitate the reaction, but hemolysis of test RBCs; less than total hemolysis
on some occasions (e.g., following effects of is graded subjectively from weak- to 3+ based on
therapy), it may be useful information. increasing color of supernatant and/or decreas-
1. Make serial dilutions (1 in 2 to 1 in 512) of the ing size of cell button).
patient’s serum in “fresh” inert serum as a
source of complement (stored serum may be Quality Control
used if correct storage conditions for comple- The complement control tube is a negative control
ment activity are applied.96 If the serum screen (no lysis). If the complement alone causes lysis, the
indicates lysis only in AS + AC, then the fresh test with the added complement is invalid. Repeat
normal serum should be acidified to pH 6.5 to the test without complement added (if not already
6.8. A control tube of the complement alone done) or with a new source of complement.
should also be set up. The patient’s 37°C only tube is a negative control
2. To 0.2 mL aliquots of each dilution, 1 drop of (no lysis). If the 37°C only tube shows lysis, repeat
10–20% group O RBCs in saline (preferably the test with the 37°C test set up prewarmed; if
buffered to pH 7–7.4). If only the enzyme- possible, include a parallel patient’s serum control
treated RBCs are hemolyzed in the screening, set up precooled at 0°C. Both the 37°C and 0°C con-
then these are substituted for the untreated RBCs. trols, set up prewarmed and precooled, respectively,
3. The tubes are mixed and incubated for 1 hour. If should be negative.
cold hemolysins are being quantitated, then the
tubes are incubated at 20°C. If warm hemolysins Interpretation
are being quantitated, then the serum dilutions 1. Hemolysis in the patient’s “test” tube (with or
and RBCs are warmed to 37°C separately for without complement added), without lysis in
approximately 10 minutes, then the cells are the control tubes, indicates a positive test. To
added. confirm that the lysis is not a false positive test
4. After incubation, mix the tubes gently (mixing is due to a monophasic lysin, place the patient’s
important) and centrifuge at 1000 RCF for 2 min- 37°C “control” tube(s) at 20°C for 60 minutes.
utes. If warm hemolysins are being measured, 2. No hemolysis in the patient’s “test” tube indi-
then the centrifugation should be at 37°C. cates a negative test.
5. Examine the supernatants for lysis and grade the
results. Notes
1. The test can also be performed by placing
Note
samples of the patient’s blood into two tubes
If the patient’s serum is already red due to in vivo
prewarmed to 37°C. One sample is left to clot at
lysis, then it is a good idea to also take into account
37°C. The other is placed immediately in melting
the size of the unlysed RBC button (i.e., compare it
ice and left undisturbed for 1 hour. It is then
with the tube containing complement only).
moved to 37°C for 30 minutes, centrifuged, and
224 Immune Hemolytic Anemias

inspected for hemolysis. This is known as the group O, P+ RBCs, and pp or Pk RBCs also
direct Donath-Landsteiner (DL) Test and is a defines the specificity of the antibody and
simple way of performing the test, but rather further confirms the diagnosis of PCH. This is
wasteful of blood, and less sensitive than the evidenced by the fact that the AGT is positive on
indirect test described above. unlysed cells remaining after performing the
2. The indication of a positive test is grossly biphasic hemolysis test.
evident in in vitro hemolysis so that presence of As a patient recovers from his illness and the
marked hemoglobinemia, which is common in antibody gradually weakens in reactivity, the
the acute stages of PCH, complicates the test. IAT may remain positive indicating that this is a
One way to observe for hemolysis when using a sensitive means of detecting the antibody. The
test serum that is already pink or red is to note specificity of the antibody as demonstrated by
the size of the RBC button after centrifugation. A the IAT will be the same as when tested by the
change in the color of the serum may not be classic bithermic hemolysis test.
evident, but only a small number of cells may
remain indicating that hemolysis has taken
place. If a change in the size of the RBC button is
not readily apparent when compared to a
control tube utilizing the same volume of RBCs, QUESTION 4: What is the specificity of the antibody
additional variations on the theme should be eluted from the patient’s RBCs and present in the patient’s
performed as follows: serum?
3. Since antibody is fixed to the RBCs in the cold It is essential to prepare an eluate from the RBCs if
phase of incubation and warming is necessary one wants to characterize (e.g., determine the
only to allow the complement cascade to specificity) of the autoantibody. If RBCs have only
proceed, the patient’s serum may be replaced by complement components demonstrable on them by
fresh normal ABO compatible serum after the the DAT, then no antibody is usually detectable in the
cold incubation. This technique is called a eluate from the RBCs. If IgG is present on the cells, it
“two-stage” DL test and is useful when the can usually be eluted by any of the many elution
patient’s serum is red before performing the test. methods described in the literature. Most of the
After incubation for 1 hour at 0°C, the super- elution data we published in 198035 were based on the
natant serum is removed from the settled RBCs ether elution method. In the 1980s, we routinely used
(centrifugation is not necessary) and replaced a xylene103 method, and in the 1990s we used an acid
with fresh normal ABO compatible serum. The elution method (Elu-Kit II, Gamma Biologicals) and
tube is then incubated at 37°C after gentle later a modification of this method following the
mixing and observed for hemolysis as described findings of some false-positive results using the origi-
above. Since a small amount of the patient’s nal method.104
serum may remain in the original tube, a control If the patient has not been transfused recently, then
tube utilizing the patient’s inactivated (heated to the eluate should contain autoantibody only; if the
56°C for 30 minutes) serum followed by replace- specificity is obvious then one can confirm the pres-
ment with normal inactivated serum will be ence of the matching antigen on the patient’s RBCs. If
necessary. a patient with hemolytic anemia has a positive DAT
4. Enzyme-treated RBCs can be used for the DL due to IgG sensitization, and the eluate shows no
test, but careful controls are necessary. On rare activity against normal cells, then an association with
occasions, the DL test is only positive with drugs might be suspected. A good example of this is
enzyme-treated RBCs.97-99 penicillin-induced hemolytic anemia where the eluate
5. It is also possible to test for the DL antibody by from DAT-positive cells will not react with untreated
the IAT, although this sometimes presents normal cells, but will react strongly with RBCs treated
difficulties because of agglutination at 0°C with penicillin (see Chapter 8). It should be empha-
caused by the DL antibody itself or by a coinci- sized that, as mentioned previously, up to 80% of
dentally occurring IgM cold agglutinin. If direct eluates prepared from random DAT+ patients are
agglutination after the cold incubation step is nonreactive and are probably due to uptake of non-
absent or weak, the RBCs may then be washed antibody proteins.
four times in ice-cold saline (to avoid elution
of the antibody) and tested with monospecific DETERMINING SPECIFICITY
anti-IgG AGS.99Although it is advisable to do
the IAT in the cold for maximal reactivity, some OF AUTOANTIBODIES
antibody may remain fixed to the cells even after
incubation at 37°C.100-102 If pp or Pk RBCs are Many interesting observations and concepts have
available, these can serve as controls, since the been derived from detailed studies of the specificity of
specificity of DL antibodies is almost always autoantibodies in AIHA (see Chapter 7). However,
anti-P. Performing the DL test against normal such exhaustive studies are not required for diagnostic
The Serologic Investigation of Autoimmune Hemolytic Anemia 225

purposes. Nevertheless, we feel it is useful (if time 3. Add 1 drop of 2% to 5% group O rr, R1R1, R2R2
allows) to perform at least limited specificity studies, RBCs to each appropriate row of tubes.
especially in patients with suspected WAIHA. This is 4. Incubate at 37°C for 15 to 60 minutes.
true because diagnostic error is avoided by 5. Wash 4 times in isotonic saline.
confirming that the patient’s warm antibody is, 6. Add AGS to button of washed RBCs.
indeed, an autoantibody. In some patients, alloanti- 7. Examine for agglutination
body(ies) may be detected in addition to autoantibody
and it is helpful to have such information available Table 6-8 shows results of an eluate that would be
since transfusion may be required. The specificity of interpreted as showing an anti-e of “relative
the autoantibody may also be of significance with specificity.” The major disadvantage of this method
regard to transfusion. Delaying specificity testing is that it assumes that the specificity is within the Rh
until a decision is made that blood is needed for trans- system, which is not always true (see Chapter 7, and
fusion (at which time the need may be urgent) is not references 105 and 106). In addition, we have often
recommended. In still other patients, the initial found that when eluates or sera showing results
impression of AIHA may not be substantiated. similar to those shown in Table 6-8 (i.e., anti-e of
Antibody found in initial screening tests of the relative specificity) are tested with more RBCs (e.g.,
patient’s serum may be alloantibody and the DAT more R2R2 cells), then the specificity pattern is
may be positive for any of the reasons listed on page different. We recommended a modification of this
213. If the patient has been transfused recently, a dis- technique35; that is, a dilution of the eluate (and/or
tinction between delayed hemolytic transfusion reac- serum) is selected that gives an approximate 1 to 2+
tions and AIHA may be difficult, and the specificity of reaction and that dilution is tested against as many
the RBC-bound antibody may be a clue to which is different phenotypes as possible; very rare RBCs
occurring (see Chapter 9). If the patient has received lacking high frequency antigens have to be included
cefotetan recently, the DAT and the apparent autoan- and possible adsorption/elution studies to define
tibody could be drug induced (see Chapter 8). many specificities.105-109 An additional advantage of
this latter approach is that the results may indicate
alloantibody (as alloantibody is commonly of higher
DETERMINING SPECIFICITY titer than autoantibody), in addition to autoantibody
OF AUTOANTIBODIES ASSOCIATED specificity. Table 6-9 illustrates a case where the
WITH WARM TYPE AUTOIMMUNE undiluted eluate reacted with all cells and the serum
HEMOLYTIC ANEMIA appeared to have relative anti-e specificity. A 1 in 64
dilution of the eluate showed relative anti-e spe-
cificity and a 1 in 4 dilution of the serum showed an
If the eluate (and/or serum) reacts with all RBCs on allo anti-K to be present in addition to the anti-“e.”
a routine panel and no specificity is obvious, the ear-
liest recommended method was to first test dilutions Differential Adsorptions and Elutions
of the eluate (and/or serum) against rr, R1R1, and The patient’s serum or eluate can be adsorbed with
R2R2 RBCs.5 If stronger reactions are obtained with RBCs of known phenotype and the adsorbed serum
some phenotypes, the results are expressed as a or eluate tested against a panel. In addition, an eluate
“relative specificity.” prepared from the adsorbing RBCs can be tested
against a panel. Such tests are time consuming and
Dilution Technique need large quantities of RBCs, but have sometimes
1. Prepare serial doubling dilutions of eluate revealed interesting specificities.107-109 Descriptions
(and/or serum) in saline from 1 in 1 to 1 in of so-called mimicking autoantibodies have made the
512 (e.g., 0.2 aliquots). interpretations of this method complex. Issitt and
2. Starting from highest dilution, transfer 2 drops coworkers109 studied 48 autoantibodies with appar-
to three 10 × 75-mm test tubes labeled rr, R1R1, ent “simple” Rh specificity (anti-e, -E, -D, -C, -Ce, -G)
R2R2. by multiple adsorption tests. The finding that 34

TABLE 6-8. ELUATE SHOWING ANTI-e OF “RELATIVE SPECIFICITY”

Dilutions of Patient’s Eluate

Phenotype of RBCs 2 4 8 16 32 64 128 256

rr 4+ 3+ 3+ 2+ 2+ 1+ 0 0
R1R1 4+ 3+ 3+ 2+ 2+ 1+ 0 0
R2R2 3+ 2+ 1+ 0 0 0 0 0
226 Immune Hemolytic Anemias

TABLE 6-9. SPECIFICITY TESTING OF ELUATE AND SERUM

IAT on Pooled Dilutions


Screening RBCs 2 4 8 16 32 64 128 256

Eluate 3+ 3+ 2+ 2+ 1+ 1+ 0 0
Serum 2+ 1+ 0 0 0 0 0 0

Selected Group O RBCs


rr rr r’r R1R1 R1R1 R1R2 R2R2 r”r”
K– K+ K– K+ K– K– K– K–

Eluate neat 3+ 3+ 3+ 3+ 3+ 3+ 3+ 3+
Eluate 1in 64 1+ 1+ 1+ 1+ 1+ 1+ 0 0
Serum neat 2+ 2+ 2+ 2+ 2+ 2+ 0 0
Serum 1in 4 0 1+ 0 1+ 0 0 0 0

(70.8%) of these antibodies could bind to RBCs DETERMINING SPECIFICITY OF


lacking the antigens that the antibodies appeared to AUTOANTIBODIES ASSOCIATED WITH COLD
define indicated that the antibodies had different AGGLUTININ SYNDROME
specificities than indicated by initial antibody
identification tests. For instance, in contrast to allo-
anti-E, autoantibodies that appeared to have anti-E
As the specificity associated with CAS is commonly
specificity could be adsorbed by E negative RBCs.
anti-I and less commonly anti-i, the specificity is
Those autoantibodies that at first appeared to be
usually obvious from the results of the cold agglutinin
directed against the Rh antigens e, E, or c, most often
titer/thermal range technique described on page 220.
had anti-Hr or anti-Hro specificity (see Chapter 7).
As all adult cells contain some i and all cord cells some
Most autoantibodies in patients with WAIHA
I, results are never clear-cut when determining
have specificity within the Rh system (see Chapter
specificity within the Ii “system.” Often, at 4°C the i
7). Using rare cells such as -D- and Rhnull, Weiner
cells will react as strongly as the I cells, and only by
and Vos107 described specificity against normal cells,
performing a titer and thermal range will the speci-
partially deleted cells, and deleted cells. Auto-
ficity become obvious (Table 6-10 shows the results of
antibodies that failed to react with Rhnull RBCs and
a typical anti-I). When cord RBCs react stronger than
had anti-U specificity have been described. It was
adult RBCs, the specificity may be anti-i but adult i
suggested that the “Rh-related” autoantibodies that
RBCs will have to be tested to confirm that these
cannot be identified as specific Rh antibodies may be
reactions are due to anti-i rather than anti-IT (see
anti-U. We examined eluates from the RBCs of eight
Chapter 7). The determination of the specificity of the
patients with AIHA using a panel of extremely rare
cold agglutinin is often only of academic interest as it
cells and cross-adsorption and elution techniques.108
is not recommended that the rare i adult blood is
We demonstrated autoantibody specificities not
obtained for these patients but that they are transfused
definable without the rare cells, and further defined
with normal I adult blood at 37°C (see Chapter 10). If
heterogeneity of the LW antigen. Autoantibodies
further confirmation of specificity is required (e.g., to
with U specificity occurred in three eluates only;
determine if antibody is possibly anti-HI, etc.), then
anti-U was always present with an antibody of
the titration/thermal range technique can be extended
another specificity. Six of the eluates contained anti-
using more cells, for example, I adult, i cord, and/or i
LW, two anti-nl, five anti-pdl, three anti-dl, and one
adult RBCs of different ABO types and Oh (Bombay)
anti-e. Adsorption and elution studies using the rare
RBCs. On rare occasions, another specificity, anti-Pr, is
Rh-positive LW-negative (Mrs. B) RBCs showed that
observed.110 This should be suspected if the I adult
anti-pdl may in fact represent anti-LW + anti-LW1
and i cord (and/or i adult) RBCs react equally at all
and that Mrs. B may represent a weak variant of LW.
phases. The Pr antigens are destroyed by proteolytic
Injection of her RBCs into guinea pigs produced an
enzymes. Thus, the cold agglutinin titer can be
anti-LW that reacted similarly to the antibody pro-
repeated comparing untreated RBCs with enzyme-
duced by injecting Rh-positive LW-positive cells. An
treated RBCs (e.g., papain-treated). Both anti-I and
analogy to the ABO was suggested: Normal Rh+
anti-i react better with enzyme-treated RBCs, but most
RBCs may represent LW1. Rh-negative LW-positive
anti-Pr will give no reaction, or much weaker re-
RBCs may represent LW2, Mrs. B may represent
actions. Other antibodies that react with antigens that
LW3, and Rhnull cells may represent the only true
are destroyed by enzyme-treatment (e.g., M and N)
LW-negative (lw).105,108
must be excluded by testing a panel of RBCs; anti-Pr
The Serologic Investigation of Autoimmune Hemolytic Anemia 227

TABLE 6-10. TYPICAL COLD AGGLUTININ TITER/THERMAL RANGE


SUGGESTING ANTI-I SPECIFICITY

4°C 25°C 30°C

Dilutions of Adult Cord Adult Cord Adult Cord


Patients’ Sera RBCs RBCs RBCs RBCs RBCs RBCs

2 4+ 4+ 4+ 1+ 1+ 0
4 4+ 4+ 4+ 1+ 1+ 0
8 4+ 4+ 4+ 1+ 1+ 0
16 4+ 4+ 3+ 1⁄2+ 1⁄2+ 0
32 4+ 4+ 2+ 0 0 0
64 4+ 4+ 1+ 0 0 0
128 4+ 3+ 1⁄2+ 0 0 0
256 3+ 3+ 0 0 0 0
512 3+ 2+ 0 0 0 0
1024 2+ 1+ 0 0 0 0
2048 1+ 0 0 0 0 0

TABLE 6-11. SUMMARY OF MOST IMPORTANT SEROLOGIC CHARACTERISTICS ASSOCIATED WITH AIHA

Type DAT Eluate Serologic Characteristics Antibody Specificity

Warm AIHA IgG and/or IgG antibody IAT positive (50% to 90% sera), agglutinating Usually within Rh system; other
complement enzyme premodified cells (90%), specificities include LW, U,
(C3dg) hemolyzing enzyme premodified cells IT, K, Kpb, K13, Ge, Jka,
(13%) Ena, and Wrb
Cold agglutinin Complement Negative Agglutinating activity up to 30°C in Usually anti-I; other
syndrome (C3dg) albumin; high titer at 4°C (usually ≥512) specificities include i, Pr,
alone Gd, and Sdx
Paroxysmal cold Complement Negative Biphasic hemolysin (i.e., sensitizing red cells Anti-P (i.e., reacts with all
hemoglobinuria (C3dg) in the cold and then hemolyzing them normal red cells except
alone when moved to 37°C) p or Pk cells)

will react with all untreated human RBCs on the panel. at 37°C, gives the expected rise in hematocrit, but if
Further confirmatory tests can be performed using this does not occur then we agree that there is an argu-
various animal RBCs and neuraminidase-treated ment for trying to obtain the blood of the rare pp phe-
RBCs110 (see Chapter 7). notype. To determine specificity, the patient’s serum
should be tested against I adult, i cord (and/or i adult),
and the rare pp and/or Pk RBCs by the DL test.
DETERMINING SPECIFICITY OF
AUTOANTIBODIES ASSOCIATED WITH
PAROXYSMAL COLD HEMOGLOBINURIA R E F E R E N C E S
1. Coombs RRA, Mourant AE, Race RR: A new test for the detec-
In our experience and in the experience of Dacie,5 the tion of weak and “incomplete” Rh agglutinins. Br J Exp Pathol
specificity of the DL antibody in PCH has always been 1945;26:255.
anti-P. There are isolated reports of other specificities.105 2. Boorman KE, Dodd BE, Loutit JF, Mollison P: Some results of
transfusion of blood to recipients with “cold” agglutinins. Br
In contrast to CAS, the specificity may be of clinical Med J 1946;i:751.
importance when transfusing patients with PCH, as 3. Loutit JF, Mollison PL: Haemolytic icterus (acholuric jaundice)
this is the only AIHA where the specificity is clear-cut congenital and acquired. J Pathol Bact 1946;58:711.
like an alloantibody. Because of this, some workers 4. Coombs RRA, Mourant AE, Race RR: In vivo iso-sensitization
of RBCs in babies with haemolytic disease. Lancet 1946;i:264.
have suggested that the rare pp RBCs have such 5. Dacie JV: The Haemolytic Anaemias, 3rd ed. vol. 3, The
better survival that it is worth trying to obtain them Auto-immune Haemolytic Anaemias. New York: Churchill
for transfusing to PCH patients with severe acute Livingstone, 1992.
hemolysis.111,112 However, since obtaining pp RBCs is 6. Dacie JV, Crookston JH, Christenson WN: “Incomplete” cold
likely to delay transfusion, which is often needed antibodies: Role of complement in sensitization to antiglobulin
serum by potentially haemolytic antibodies. Br J Haematol
urgently in PCH patients, we advise using common P 1957;3:77.
positive blood in such instances. Our limited experi- 7. Petz LD, Garratty G: Antiglobulin sera—past, present and
ence has been that common P positive blood, if given future. Transfusion 1978;18:257–268.
228 Immune Hemolytic Anemias

8. Garratty G: The significance of complement in immunohema- 34. Gilliland BC: Coombs-negative immune hemolytic anemia.
tology. Crit Rev Clin Lab Sci 1985;20:25. Semin Hematol 1976;13:267–275.
9. Freedman J: Complement in transfusion medicine. In: Garratty 35. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias,
G, ed. Immunobiology of Transfusion Medicine. New York: 1st ed. New York: Churchill Livingstone, 1980.
Dekker, 1994:403–434. 36. Merry AH, Thomsen EE, Rawlinson VI, Stratton F: A quantita-
10. Issitt PD, Anstee DJ: Applied Blood Group Serology, 4th ed. tive antiglobulin test for IgG for use in blood transfusion serol-
Durham, NC: Montgomery Scientific Publications, 1998. ogy. Clin Lab Haematol 1982;4:393–402.
11. Garratty G, Petz LD: An evaluation of commercial antiglobu- 37. Jeje MO, Blajchman MA, Steeves K, Horsewood P, Kelton JG:
lin sera with particular reference to their anticomplement Quantification of RBC-associated IgG using an immunoradio-
properties. Transfusion 1971;11:79–88. metric assay. Transfusion 1984;24:473–476.
12. Burkart P, Rosenfield RE, Hsu TCS, et al.: Instrumented 38. Kay MMB: Mechanism of removal of senescent cells by
PVP-augmented antiglobulin tests. I. Detection of allogeneic human macrophages in situ. Proc Natl Acad Sci USA 1975;
antibodies coating other normal erythrocytes. Vox Sang 72:3521–3525.
1974;26:280. 39. Alderman EM, Fudenberg HH, Lovins RE: Binding of
13. Mollison PL, Engelfriet CP, Contreras M: Blood Transfusion in immunoglobulin classes to subpopulations of human red
Clinical Medicine, 10th ed. Oxford: Blackwell Scientific, 1997. blood cells separated by density-gradient centrifugation.
14. Worlledge SM, Blajchman MA: The autoimmune haemolytic Blood 1980;55:817–822.
anemias. Br J Haematol 1972;23(Suppl.):61. 40. Szymanski IO, Odgren PR, Fortier NL, et al: Red blood cell
15. Sokol RJ, Booker DJ, Stamps R: The pathology of autoimmune associated IgG in normal and pathologic states. Blood
haemolytic anaemia. J Clin Pathol 1992;45:1047–1052. 1980;55:48–54.
16. Garratty G: Autoimmune hemolytic anemias. In: Garratty G, 41. Freedman J: Membrane-bound immunoglobulins and comple-
(ed). Immunobiology of Transfusion Medicine. New York: ment components on young and old RBCs. Transfusion
Dekker, 1994:523–551. 1984;24:477–481.
17. Petz LD, Garratty G: Laboratory correlations in immune 42. Kay MMB: Localization of senescent cell antigen on band 3.
hemolytic anemias. In: Vyas GN, Sites DP, Brecher G, (eds). Proc Natl Acad Sci USA 1984;81:5753–5757.
Laboratory Diagnosis of Immunologic Disorders. New York: 43. Lutz HU, Flepp R, Stringaro-Wipf G: Naturally occurring
Grune & Stratton, 1975. autoantibodies to exoplasmic and cryptic regions of band 3
18. Chaplin H, Jr: Clinical usefulness of specific antiglobulin protein, the major integral membrane protein of human red
reagents in autoimmune hemolytic anemias. In: Brown EB, blood cells. J Immunol 1984;133:2610–2618.
(ed). Progress in Hematology, vol. VIII. New York: Grune & 44. Low PS, Waugh SM, Zinke K, et al: The role of hemoglobin
Stratton, 1973. denaturation and band 3 clustering in red blood cell aging.
19. Dacie JV: Occurrences in normal human sera of “incomplete” Science 1985;227:531–533.
forms of “cold” autoantibodies. Nature (Lond) 1950;166:36. 45. Graham HA, Davies DM, Jr, Brower CE: Detection of C3b and
20. Garratty G: The clinical significance (and insignificance) of C3d on RBC membranes. International Symposium on the
red-cell-bound IgG and complement. In: Wallace ME, Levitt JS, Nature and Significance of Complement Activation. Raritan
(eds). Current Applications and Interpretation of the Direct Ortho Research Institute of Medical Sciences, 1976:107–111.
Antiglobulin Test. Arlington, VA: American Association of 46. Rosenfield RE, Jagathambal K: Antigenic determinants of C3
Blood Banks, 1988:1–24. and C4 complement components on washed erythrocytes from
21. Garratty G: The significance of IgG on the RBC surface. normal persons. Transfusion 1978;18:517–523.
Transfus Med Rev 1987;1:47–57. 47. Freedman J, Barefoot C: Red blood cell-bound C3d in normal
22. Toy PTCY, Chin CA, Reid ME, et al: Factors associated with subjects and in random hospital patients. Transfusion
positive direct antiglobulin tests in pretransfusion patients: A 1982;22:511–514.
case-control study. Vox Sang 1975;49:216-220. 48. Chaplin H, Nasongkla M, Monroe MC: Quantification of red
23. Heddle NM, Kelton JG, Turchyn KL, et al: Hypergamma- blood cell-bound C3d in normal subjects and random hospi-
globulinemia can be associated with a positive direct antiglob- talized patients. Br J Haematol 1981;48:69–78.
ulin test, a nonreactive eluate, and no evidence of hemolysis. 49. Merry AH, Thomsen EE, Rawlinson VI, Stratton F: The
Transfusion 1988;28:29–33. quantification of C3 fragments on erythrocytes: Estimation of
24. Garratty G: Problems associated with passively transfused C3 fragments on normal cells, acquired haemolytic anaemia
blood group alloantibodies. Am J Clin Pathol 1998;109:769–777. cases and correlation with agglutination of sensitized cells.
25. Jandl JH: The agglutination and sequestration of immature Clin Lab Haematol 1983;5:387–397.
RBCs. J Lab Clin Med 1960;55:663. 50. Freedman J: Membrane-bound imunoglobulins and comple-
26. Sutherland DA, Eisentraut AM, McCall MS: The direct ment components on young and old RBCs. Transfusion
Coombs test and reticulocytes. Br J Haematol 1963;9:68–76. 1984;24:477–481.
27. Worlledge SM: The interpretation of a positive direct anti- 51. Szymanski IO, Odgren PR, Valeri CR: Relationship between
globulin test. Br J Haematol 1978;39:157. the third component of human complement (C3) bound to
28. Stratton F, Renton PH: Effect of crystalloid solutions pre- stored preserved erythrocytes and their viability in vivo. Vox
pared in glass bottles on human RBCs. Nature (London) Sang 1985;49:34–41.
1955;175:722. 52. Salama A, Mueller-Eckhardt C: Binding of fluid phase C3b to
29. Jandl JH, Simmons RL: The agglutination and sensitization of nonsensitized bystander human RBCs. Transfusion
RBCs by metallic cations: Interactions between multivalent 1985;25:528–534.
metals and the RBC membrane. Br J Haemat 1957;3:19. 53. Siegel I, Liu TL, Gleicher N: The red-cell immune system.
30. Sturgeon P, Smith LE, Chun HMT, Hurvitz CH, Garratty G, Lancet 1981;ii:556–559.
Goldfinger D: Autoimmune hemolytic anemia associated exclu- 54. Weiner W: “Coombs positive” “normal” people. Proceedings
sively with IgA of Rh specificity. Transfusion 1979;19:324–328. of the Tenth Congress of the International Society of Blood
31. Casey FM, Dodd BE, Lincoln PJ: A study of the characteristics Transfusion, Stockholm, 1964:34–39.
of certain Rh antibodies preferentially detectable by enzyme 55. Habibi B, Muller A, Lelong F, et al: Auto-immunisation ery-
technique. Vox Sang 1972;23:493–507. throcytaire dans la population “normale.” Nouvelle Presse Ed
32. Hughes-Jones NC, Polley MJ, Telford R, Gardner B, 1980;43:3253–3257.
Kleinschmidt G: Optimal conditions for detecting blood group 56. Gorst DW, Rawlinson VI, Merry AH, et al: Positive direct
antibodies by the antiglobulin test. Vox Sang 1964;9:385. antiglobulin test in normal individuals. Vox Sang 1980;38:99–105.
33. Dupuy ME, Elliot M, Masouredis SP: Relationship between 57. Allan J, Garratty G: Positive direct antiglobulin tests in normal
RBC bound antibody and agglutination in the antiglobulin blood donors. Proceedings of the International Society of
reaction. Vox Sang 1964;9:40. Blood Transfusion, Montreal, 1980:150 (Abstr).
The Serologic Investigation of Autoimmune Hemolytic Anemia 229

58. Bareford D, Longster G, Gilks L, et al: Follow-up of normal 84. Holtz G, Mantel W, Buck W: The inhibition of antigen-
individuals with a positive antiglobulin test. Scand J Haematol antibody reactions by chloroquine and its mechanism of
1985;35:348-53. action. Z Immun Forsch Bd 1973;146:145–157.
59. Pirofsky B: Autoimmunization and the Autoimmune 85. Mantel W, Holtz G: Characterisation of autoantibodies to ery-
Hemolytic Anemias. Baltimore: Williams & Wilkins, 1969. throcytes in autoimmune haemolytic anaemia by chloroquine.
60. Freedman J: False-positive antiglobulin tests in healthy Vox Sang 1976;30:453463.
subjects and in hospital patients. J Clin Pathol 1979; 86. Edwards JM, Moulds JJ, Judd WJ: Chloroquine dissociation of
32:1014–1018. antigen-antibody complexes. Transfusion 1982;22:59–61.
61. Bohnen RF, Ultmann JE, Gorman JG, Farhangi M, Scudder J: 87. Sassetti R, Nichols D: Decreased antigenic reactivity caused by
The direct Coomb’s test: Its clinical significance. Ann Intern chloroquine. Transfusion 1982;22:537–538.
Med 1968;68:19–32. 88. Mallory D, Reid M: Misleading effects of chloroquine (letter).
62. Lau P, Haesler WE, Wurzel HA: Positive direct antiglobulin Transfusion 1984;24:412.
reaction in a patient population. Am J Clin Pathol 1975; 89. McShane K, Cronwall S: Chloroquine reduces antigen strength
65:368–375. (letter). Transfusion 1985;25:83.
63. Judd WJ, Butch SH, Oberman HA, Steiner EA, Bauer RC: The 90. Reid ME: Autoagglutination dispersal utilizing sulphydryl
evaluation of a positive direct antiglobulin test in pretransfu- compounds. Transfusion 1998;18:353–355.
sion testing. Transfusion 1980;20:17–23. 91. Merry AH, Thomson EE, Rawlinson VI, et al: Quantification of
64. Judd WJ, Barnes BA, Steiner EA, Oberman HA, Averill DB, IgG on erythrocytes: correlation of number of IgG molecules
Butch SH: The evaluation of a positive direct antiglobulin test per cell with the strength of the direct and indirect antiglobu-
(autocontrol) in pretransfusion testing revisited. Transfusion lin tests. Vox Sang 1984;47:73–81.
1986;26:220–224. 92. Kumpel BM: A simple non-isotopic method for the quanti-
65. Huh YO, Lichtiger B: Evaluation of a positive autologous fication of RBC-bound immunoglobulin. Vox Sang 1990;
control in pretransfusion testing. Am J Clin Pathol 1985; 59:34–38.
84:632–636. 93. Dubarry M, Charron C, Habibi B, et al: Quantification of
66. Toy PTCY, Reid M, Burns M: Positive direct antiglobulin test immunoglobulin classes and subclasses of autoantibodies
associated with hyperglobulinemia in acquired immuno- bound to RBCs in patients with and without hemolysis.
deficiency sydnrome (AIDS). Am J Hematol 1985;19:145–150. Transfusion 1993;33:466–471.
67. Chaplin H, Nasongkla M, Monroe MC: Quantification of red 94. Garratty G, Nance SJ: Correlation between in vivo hemolysis
blood cell-bound C3d in normal subjects and random hospi- and the amount of red cell-bound IgG measured by flow
talized patients. Br J Haematol 1981;48:69–78. cytometry. Transfusion 1990;30:617–621.
68. Freedman J, Ho M, Barefoot C: Red blood cell-bound C3d in 95. Garratty G, Petz LD, Hoops JK: The correlation of cold agglu-
selected hospital patients. Transfusion 1982;22:515–520. tinin titrations in saline and albumin with haemolytic
69. Garratty G, Arndt PA: Application of flow cytofluorometry to anaemia. Br J Haemat 1977;35:587–595.
red blood cell immunology. Cytometry 1999;38:259–267. 96. Garratty G: The effect of storage and heparin on serum com-
70. Hoppe PA: Background for current federal requirements for plement activity with particular reference to the detection of
antiglobulin reagents in the United States. In: Chaplin H, Jr blood group antibodies. Am J Clin Pathol 1970;54:531–538.
(ed). Immune Hemolytic Anemias. New York: Churchill 97. Heddle NM: Acute paroxysmal cold hemoglobinuria. Transfus
Livingstone, 1984:209. Med Rev 1989;III:219–229.
71. Garratty G, Petz LD: The significance of RBC bound comple- 98. Sokol RJ, Booker DJ, Stamps R: Paroxysmal cold hemoglobin-
ment components in development of standards and quality uria and the elusive Donath-Landsteiner antibody.
assurance for the anti-complement components of antiglobu- Immunohematology 1998;14:109–112.
lin sera. Transfusion 1976;16:297–306. 99. Worlledge SM, Rousso C: Studies on the serology of paroxys-
72. Fruitstone MJ: C3b-sensitized erythrocytes (letter). Transfusion mal cold haemoglobinuria (PCH), with special reference to its
1978;18:125. relationship with the P blood group system. Vox Sang
73. Chaplin H, Friedman J, Massay A, Monroe MC: Charac- 1965;10:293–298.
terization of red blood cells strongly coated in vitro by C3 via 100. Ries CA, Garratty G, Petz LD, et al: Paroxysmal cold hemoglo-
the alternative pathway. Transfusion 1980;20:256–262. binuria: report of a case with an exceptionally high thermal
74. Moore JA, Chaplin H: Anti-C3d antiglobulin reagents. range Donath-Lansteiner antibody. Blood 1971;38:491–499.
Transfusion 1974;14:407–415. 101. Lindgren S, Zimmerman S, Gibbs F, Garratty G: An unusual
75. Garratty G, Arndt PA: Evaluation of IgG sensitized RBCs used Donath-Landsteiner antibody detectable at 37°C by the
for controlling antiglobulin tests. Transfusion 1999;39:104S antiglobulin test. Transfusion 1985;25:142–144.
(abstr). 102. Nordhagen R: Two cases of paroxysmal cold hemoglobinuria
76. Garratty G: Low affinity autoantibodies: A cause of false nega- with a Donath-Landsteiner antibody reactive by the indirect
tive direct antiglobulin tests. Transfus Today 1991;12:3–4. antiglobulin test using anti-IgG (letter). Transfusion 1991;31:190.
77. Postoway N, Garratty G: Standardization of IgG subclass anti- 103. Bueno R, Garratty G, Postoway N: Elution of antibody from
serums for use with sensitized RBCs. Transfusion 1983; RBCs using xylene—a superior method. Transfusion 1981;
23:398–400. 21:157–162.
78. Garratty G, Postoway N, Nance S, Brunt D: Spontaneous 104. Leger RM, Arndt PA, Ciesielski DJ, Garratty G: False-positive
agglutination of RBCs with a positive direct antiglobulin test eluate reactivity due to the low-ionic wash solution used with
in various media. Transfusion 1984;24:214–217. commercial acid-elution kits. Transfusion 1998;38:565–572.
79. Rodberg K, Tsuneta R, Garratty G: Discrepant Rh phenotyping 105. Garratty G: Target antigens for red-cell-bound autoantibodies.
results when testing IgG-sensitized RBCs with monoclonal Rh In: Nance SJ, (ed). Clinical and Basic Science Aspects of Immu-
reagents. Transfusion 1995;35(Suppl.):67S. nohematology. Arlington, VA: American Association of Blood
80. Louie JE, Jiang AF, Zaroulis CG: Preparation of intact anti- Banks, 1991:33–72.
body-free red blood cells in autoimmune hemolytic anemia. 106. Garratty G: Specificity of autoantibodies reacting optimally at
Transfusion 1986;26:550 (Abstr). 37°C. Immunohematology 1999;15:24–40.
81. Rekvig OP, Hannestad K: Acid elution of blood group anti- 107. Weiner W, Vos GH: Serology of acquired hemolytic anemias.
bodies from intact erythrocytes. Vox Sang 1977;33:280–285. Blood 1963;22:606–613.
82. des Roziers NB, Squalli S: Removing IgG antibodies from 108. Vos GH, Petz LD, Garratty G, Fudenberg HH: Autoantibodies
intact red cells: Comparison of acid and EDTA, heat, and in acquired hemolytic anemia with special group reference to
chloroquine elution methods. Transfusion 1997;37:497–501. the LW system. Blood 1973;42:445–453.
83. Byrne PC: Use of a modified acid/EDTA elution technique. 109. Issitt PD, Zellner DC, Rolih SD, Duckett JB: Autoantibodies
Immunohematology 1991;7:46–47. mimicking alloantibodies. Transfusion 1977;17:531–538.
230 Immune Hemolytic Anemias

110. Roelcke D: Cold agglutination. Transfus Med Rev 1989; 112. Rosenfield RE, Jagathambal K: Transfusion therapy for
3:140–166. autoimmune hemolytic anemia. Semin Haematol 1976;13:311.
111. Rausen AR, LeVine R, Hsu TCS, Rosenfield RE: Compatible
transfusion therapy for paroxysmal cold hemoglobinuria.
Pediatrics 1975;55:275–278.
C H A P T E R 7

Specificity of
Autoantibodies

Up to 1953, autoantibodies ent times). Wiener and coworkers8 suggested that


were generally considered to 37°C-reactive (“warm”) autoantibodies might be
be “nonspecific”; that is, they directed against the “nucleus of the Rh-Hr substance.”
reacted with all human red Pirofsky and Pratt, in 1966,9 compared the reactions of
blood cells (RBCs) tested, alloanti-Rh and “warm” autoantibodies with RBCs
although some variation in from a large variety of primates and nonprimates and
reaction had been observed.1,2 essentially agreed with the findings of Wiener and
The first convincing data sug- coworkers8; neither Rh alloantibodies nor autoantibod-
gesting that autoantibody ies reacted with nonprimate RBCs or certain primates
specificity could be defined (e.g., tree shrew, fulvus lemur, and woolly monkey),
were presented by Race, Sanger, and Selwyn in 1951.3 but both reacted with other primates (ringtail lemur,
squirrel monkey, celebes ape, rhesus monkey, baboon,
SPECIFICITIES ASSOCIATED WITH and chimpanzee). The authors concluded that the data
strongly suggested that warm autoantibodies have
WARM-ANTIBODY TYPE AUTOIMMUNE specificity for Rh antigens.8,9 In the next decade, many
HEMOLYTIC ANEMIA other workers reported on autoantibody specificities
associated with WAIHA.10
Rh and LW Specificity. Using RBCs from the With the exception of one anti-B,11 one anti-Jka,11
recently discovered rare phenotype –D–, Race and and two examples of anti-K,12,13 all of the specificities
coworkers3 showed that the “nonspecific” autoanti- reported during this time were associated with Rh.
bodies could be subdivided into two groups; one group Autoanti-e was the most common reported specificity;
that reacted with –D– RBCs, and the other that did not; it has been pointed out that the reported relative inci-
this suggested some Rh specificity. The first example of dence of different specific Rh autoantibodies corre-
a clearly specific autoantibody was an autoanti-e in an sponds well with the incidence of Rh antigens in the
R1R1 patient described by Weiner and coworkers in population (i.e., e is present on the RBCs of approx-
19534; others soon followed. Holländer5 described an imately 98% of the population).10 In 1963 Weiner and
autoanti-c in an R1r patient and showed that an addi- Vos14 expanded the work of Race and coworkers,3
tional “nonspecific” element was also present in the who used RBCs of the rare phenotype –D– to demon-
eluate made from the patient’s RBCs. In 1953 and 1954, strate “Rh” specificity. Weiner and Vos14 studied 60
Dacie6 and Dacie and Cutbush7 reported specificities RBC eluates that were initially thought to be non-
on 10 patients with warm-antibody type autoimmune specific; these eluates were tested against RBCs of
hemolytic anemia (WAIHA): one R1r patient had common phenotypes and rare phenotypes such as
autoanti-C+e; the other nine patients had “nonspecific” –D–, cD–, and the recently discovered Rhnull.
autoantibodies, but three also had clearly demonstrable Adsorptions, elutions (i.e., eluates made from RBCs
autoanti-e and one patient autoanti-e and -D (at differ- incubated in vitro with eluates prepared from patients’
231
232 Immune Hemolytic Anemias

TABLE 7-1. AUTOANTIBODIES IN ACQUIRED HEMOLYTIC ANEMIA


AND THEIR SEROLOGIC REACTIVITY FOR SELECTED RED BLOOD
CELLS (RBCs)

Anti-nl Anti-pdl Anti-dl “Antigens”

RBCs with common


(“normal”) phenotypes + + + nl, pdl, dl
“Partially deleted” cells
(–D–/–D– or cD–/cD–) – + + pdl, dl
“Fully deleted” cells
[– – –/– – – (Rhnull)] – – + dl

RBCs sensitized in vivo), and titrations were per- Leddy and coworkers19 studied eluates from RBCs
formed, using saline, albumin, enzymes, and indirect of 46 patients with WAIHA. They used a panel of rare
antiglobulin tests (IATs). Fifty-three percent of the human RBCs including Rhnull, –D–, LW negative, and
eluates failed to react with Rhnull RBCs, and 18% Ko RBCs as well as monkey RBCs (rhesus and stump
reacted weaker than with RBCs of common pheno- tailed monkeys). Sixteen of 20 eluates derived from
types. Thus, approximately 70% of the eluates patients whose RBCs were sensitized with both IgG
appeared to be reacting with Rh antigens. The pat- and complement reacted with all RBCs tested.
terns of reactivities suggested three different autoanti- Conversely, 12 of 26 eluates from “IgG only” RBCs
body types that could be present separately or in did not react with Rhnull RBCs, suggesting “Rh”
combination. They were termed anti-nl (normal), specificity. Leddy and coworkers19 found four pat-
anti-pdl (partially deleted), and anti-dl (deleted) terns of reactions that suggested a working
(Table 7-1). The nl determinants were said to be classification: Group I reacted with RBCs of common
present on RBCs of common phenotypes but not –D–, phenotype but were negative with Rhnull and monkey
Dc–, or Rhnull; the pdl determinants present on RBCs RBCs, and no complement was fixed in vitro; group II
of common phenotypes, –D–, and Dc–, but not Rhnull; reacted with RBCs of common phenotype and weaker
and the dl determinants present on RBCs of common with Rhnull, about 25% reacted with monkey RBCs;
phenotypes, –D–, Dc–, and Rhnull. Thus, if an autoan- group III reacted with RBCs of common phenotype
tibody reacts with all RBCs of common phenotype but and Rhnull RBCs equally but did not react with
not “partially deleted” or “fully deleted” RBCs, the monkey RBCs; group IV reacted equally with RBCs of
autoantibody would be classified as having anti-nl normal phenotypes, Rhnull, and monkey RBCs.
specificity. If the autoantibody reacts with all RBCs, it Vos and coworkers15 studied 24 sera and RBC
may be anti-dl but selective adsorptions may reveal a eluates from WAIHA. They showed that the IAT using
mixture of anti-dl + pdl, anti-dl + nl, or anti-dl + pdl + untreated RBCs often showed different specificities
nl. In other words, adsorption of a “nonspecific” than tests using ficin. As shown by others, they
autoantibody with Rhnull RBCs may reveal an anti-
body only reacting with RBCs of common phenotype
(Table 7-2). We confirmed these findings in our own
laboratory and collaborated with Vos to extend the TABLE 7-2. ANTI-nl, -pdl, AND -dl AUTOANTIBODIES
original observations.15-17 It should be pointed out MAY HAVE A WELL-DEFINED BLOOD GROUP
that autoanti-nl, -pdl, and -dl show patterns similar to SPECIFICITY IF TESTED AGAINST RED BLOOD CELLS
antibodies of the later-defined specificities shown in (RBCs) OF RARE PHENOTYPES (E.G., NULL
Table 7-2, and may be identical specificities. PHENOTYPES OTHER THAN RHnull)
In a detailed review in 1969, Dacie and Worlledge18
reviewed results on their series of AIHA patients seen Anti-nl* Anti-pdl* Anti-dl*
over a 20-year period. Specificity testing of eluates
-Hr -Rh29 -Ena
from 98 patients with WAIHA were reported. When -Hr0 -LWa -Wrb
tested against a panel of RBCs with common pheno- -Rh34 -LWab -Dib
types, approximately 30% showed obvious Rh -U -Ge
specificity (23 anti-e, 4 anti-c, 1 anti-C + D, 1 anti-D + -Kpb
-K13
e). When –D– RBCs were added to the panel, an addi- -Sc1/Sc3
tional 17% could be classified as “Rh” specific (47% of -Vel
the total) and if Rhnull RBCs were added, then an addi- -AnWj
tional 5% could be added, giving a total of 52%
* Anti-nl, reacts with RBCs of common phenotype, but not –D– or Rhnull RBCs;
showing some “Rh” specificity; results were very Anti-pdl, reacts with RBCs of common phenotype and –D–, but not Rhnull RBCs;
similar to those of Weiner and Vos.14 Anti-dl, reacts with RBCs of common phenotype, –D–, and Rhnull RBCs.
Specificity of Autoantibodies 233

reported that the eluates from RBCs sensitized only such as anti-nl, -pdl, -dl, and anti-Wrb (see later) in 23
with IgG showed clearer specificity than the “IgG+ of 87 (26%) cases. Overall, 74% of the autoantibodies
complement” group; 72% of the “IgG only” group did reacted as well with –D– and Rhnull RBCs as with
not react with Rhnull RBCs, in contrast to 100% of the common phenotypes. This study also confirmed
“IgG+ complement” group reacting with Rhnull RBCs earlier studies16 in terms of the immunoglobulin class
in addition to common phenotypes. Additional of the autoantibodies and the fact that complement
studies using RBC eluates revealed that no direct cor- fixation in vivo generally occurs only when autoanti-
relation could be established between the presence of bodies with complex specificity, such as anti-pdl or
complement components on the patient’s RBCs, as anti-dl, are produced.
determined by direct antiglobulin testing (DAT), and In 1967, Celano and Levine24 demonstrated anti-LW
the intensity of the IAT using anti-IgG serum. On the in all six cases of WAIHA that they studied. All six
other hand, the simultaneous presence of anti-nl and showed “nonspecific” reactions when tested against a
anti-dl autoantibodies in eluates was often associated panel of RBCs of common phenotype, but when Rhnull
with the presence of complement on the RBCs. They and Rh+LW– and/or Rh–LW– RBCs were included,
suggested that the presence of complement compo- one serum showed clear anti-LW specificity. Further
nents in association with anti-nl and anti-dl may be adsorption and elution studies on the other five cases
analogous to complement fixation by multiple Rh revealed the presence of anti-LW, together with a
alloantibodies as described by Rosse.20 “nonspecific” element. In 1973, we collaborated with
In a later publication, Vos and coworkers16 showed Vos to study, in depth, eight eluates made from RBCs
that the presence of complement on RBCs, in addition of patients with WAIHA.17 We used a panel of
to IgG, seemed to be associated exclusively with RBCs containing very rare phenotypes (e.g., LW+U–,
autoantibodies of multiple immunoglobulin classes, as LW–U+, Rhnull LW–U+, Rhnull LW–U–) and per-
well as multiple RBC specificities. Vos postulated that formed selected adsorption and elution studies. We
the variability encountered in multiple antibodies may were able to demonstrate specificities not definable
reflect continuous differences in the inciting stimulus without the use of some of these exotic phenotypes.
resulting from altered configuration of red cell anti- Three of the eight (38%) eluates were shown to
genic determinants. Assuming that trapping and pro- contain anti-U and six of eight (75%) contained anti-
cessing of antigen are prerequisites for the induction of LW. The anti-LW and anti-U were always detected
antibody response, it was postulated that an adequate together with other antibody specificities. The
concentration of anti-nl on the RBCs might block sites definition of these antibodies was only possible
inhibiting synthesis of antibody, but not necessarily the because we had access to the only known examples of
enhancement of antibody synthesis for other Rhnull LW–U+ and Rh+LW–; this also explains why
unblocked antigens on the immunizing RBCs (e.g., we detected anti-U in 38% of the eluates compared
anti-pdl and anti-dl). The suggestion that multiple anti- with only 6% reported by Marsh and coworkers.25
body and immunoglobulin formation follows a prede- Other rare phenotypes such as the only example of
termined sequence of development as a consequence of Wr(b-) and Ko RBCs have also revealed new
variability in antigen presentation is not in full agree- specificities among autoantibodies.
ment with the idea that the disease primarily results Of particular interest in the Vos and coworkers17
from an aberration of the immune mechanism. The study was the observation that adsorption and elution
authors suggest that WAIHA associated with Rh anti- studies, including the use of Rh+LW– RBCs, showed
bodies may result from a defect in the structural com- that antibodies previously classified as anti-pdl (anti-
position of the genome that is rejected by a normal bodies reacting with common phenotypes and –D–
immune mechanism. The subsequent development of but not Rhnull phenotypes) appeared to be anti-LW +
additional specificities to other RBC antigens involving LW1. We made the analogy to the ABO system, where
multiple immunoglobulin classes does not necessarily anti-A can be shown to be anti-A + A1 by adsorption
indicate the establishment of an aberrant immune with A2 RBCs.17 We suggested that Rh+LW+ RBCs
apparatus. Sokol and coworkers21 also reported a cor- should be classified as LW1, Rh–LW+ RBCs as LW2,
relation of RBC-bound multiple immunoglobulin and the rare Rh+LW– RBCs as LW3.17 In 1981 a new
classes with severity of hemolytic anemia. antibody (anti-Nea) was found to be part of the LW
In 1975 Dacie22 reported further data on 121 patients system.26 Anti-Nea was renamed anti-LWb and the old
with WAIHA. “Rh” specificity could be demonstrated anti-LW was renamed anti-LWa. Sistonen and
in 68% of the patients and was as high as 83% if only Tippett26 suggested that LW1 (i.e., D+LW+) are of the
patients with IgG but no complement on their RBCs genotype LWaLWa or LWaLW; LW2 (D–LW+) are
were considered; only 37% of patients with IgG and LWaLWb; LW3 are LWbLWb or LWbLW; and LW4 are
complement on their RBCs showed “Rh” specificity. LWLW (the only true “LW negative”). The antibody
In an extensive study published in 1976, Issitt and LW4 individuals make is an inseparable anti-LWab.
coworkers23 found “obvious” anti-Rh specificity Thus, the autoanti-LW described previously were
(specificities seen included anti-D, -C, -E, -c, -e, -f, -Ce, probably anti-LWa or anti-LWab.17,27 Transient anti-
and anti-G) to be present alone in 3 of 87 cases (3.5%), LW has been described in individuals who produced
together with autoantibodies of other specificities, anti-LW at a time they typed as LW– but were later
234 Immune Hemolytic Anemias

found to be LW+; these will be described later in the RBCs. In two of these cases, adsorption and elution
section on “mimicking” antibodies. studies resulted in the separation of anti-e and anti-U
It should be noted that many of the above studies with an additional “nonspecific” component reacting
used eluates rather than sera from patients with with Rhnull RBCs. Adsorption and elution studies in a
“warm” autoantibodies to define specificity. Auto- third case yielded anti-U and “nonspecific” antibody
antibodies present in sera usually show “cleaner” only. Thus, in 50 eluates, 3 (6%) showed anti-U
specificity than those eluted from RBCs. Another specificity. Other workers since have also demon-
point to notice is that some study populations were strated autoanti-U specificity associated with WAIHA,
composed of only patients with hemolytic anemia, and in patients with a positive DATs and no hemolytic
and others included patients/donors with “warm” anemia.27 In our study with Vos17 described previ-
autoantibodies but no hemolytic anemia. The clarity ously, we used the only Rhnull U+ red cell sample so
of the specificity may vary within these two popula- far described, in addition to an Rhnull U– sample, to
tions. Unfortunately, some studies did not define the study eight selected RBC eluates from patients with
clinical status of their populations. A further differ- WAIHA. We were able to demonstrate autoantibodies
ence in clarity of specificity is observed if patients are with U specificity in three of the eight eluates (38%)
subdivided into those with only IgG on their RBCs or but they were always present with autoantibodies of
whether other proteins are present (e.g., complement). other specificity (e.g., anti-LW, -nl, -pdl, -dl, and
For instance, Dacie and Worlledge18 found that, using anti-e). Without the rare Rhnull RBCs, it would have
a panel of RBCs of common phenotypes, 38% of “IgG been very difficult to demonstrate the presence of the
alone” DATs were associated with Rh specificity in anti-U and this probably accounts for the much higher
contrast to only 10% in the “IgG and complement” incidence of autoanti-U detected compared with other
population. When RBCs of the –D– and Rhnull pheno- series (e.g., Marsh and coworkers25).
types were included, 68% of “IgG alone” group In 1972, Worlledge31 reported that approximately
showed “Rh” specificity in contrast to 14% of the “IgG 33% of anti-dl autoantibodies failed to react with En(a–)
and complement” group. Eyster and Jenkins28 studied RBCs. She also noted that those anti-dl autoantibodies
eluates from 37 patients. One eluate showed anti-e most likely to fail to react with En(a–) RBCs were the
specificity, another failed to react with some cells but same ones that reacted less strongly with enzyme-
no specificity was obvious; the other 35 reacted with treated RBCs than untreated RBCs.31 A study from the
all RBCs tested; of the 35 eluates that reacted with all same department in a later report found 44% of 23
RBCs, 6 of 12 eluates from RBCs having an “IgG only” “nonspecific” eluates to react weaker, or not at all, with
positive DAT showed “Rh” specificity, whereas only 6 En(a–) RBCs.22 In 1975, Goldfinger and coworkers32
of 23 “IgG and complement” eluates showed reported a case of WAIHA associated with an autoanti-
specificity. They also confirmed the experiments of body of Wrb specificity. The antibody was initially
Weiner and Vos14 by showing that if the “nonspecific” noticed to react with all RBCs tested (including many
eluates were adsorbed with Rhnull RBCs, then the RBCs lacking high-frequency antigens) but gave
adsorbed serum sometimes demonstrated relative Rh weaker reactions with heterozygous Wr(a+b+) RBCs
specificity with RBCs of common phenotypes. than with the common Wr(a–b+) RBCs. The specificity
was confirmed by finding a negative reaction against
the only known example of Wr(a+b–) RBCs. Issitt and
SPECIFICITIES ASSOCIATED coworkers23 found that the antibody reacted weakly
WITH GLYCOPHORINS with En(a–) RBCs. The En(a–) RBCs, when tested with
the only known example of anti-Wrb, were found to
The first undisputed obvious specificity, other than type Wr(b–). It was suggested that the anti-Ena
Rh, in patients with WAIHA was described in 1971 specificity reported earlier by Worlledge31 may perhaps
when Nugent and coworkers29 described a patient have been anti-Wrb instead. The only known example
with an autoantibody with U blood group specificity. of Wr(a+b–) RBCs typed as En(a+), thus anti-Wrb and
Because of the observations of Schmidt and cowork- anti-Ena do not have the same specificity.
ers30 that Rhnull RBCs also have aberrant U antigen, In a detailed study, Issitt and coworkers reported on
together with some degree of abnormality of the Ss anti-Wrb and other autoantibodies responsible for pos-
antigens, Marsh and coworkers25 thought autoanti- itive DATs in 150 individuals.23 Of 87 patients with
bodies that were being called “Rh” specific because AIHA, 64 eluates (73.6%) had autoantibodies reacting
they failed to react with Rhnull RBCs should be reex- with all RBCs including Rhnull. Of these 64 anti-dl
amined. They studied eluates from 50 patients that autoantibodies, 34 contained autoanti-Wrb (53.1%). Of
reacted with all RBCs of common phenotypes; 24 of 33 patients being treated with methyldopa who had
these eluates reacted significantly weaker with Rhnull developed positive DATs, 23 had anti-dl autoanti-
RBCs and in some of these cases, well-defined bodies (69.7%), 4 (17.4%) of which contained
specificity for known Rh antigens could be demon- autoanti-Wrb. Of 30 hematologically normal donors
strated (e.g., anti-e). Three of the 24 cases showed with positive DATs, 23 (76.7%) eluates contained
autoantibodies that reacted with all RBCs of common anti-dl autoantibodies and 8 (34.8%) eluates contained
phenotype but more weakly with both U− and Rhnull autoanti-Wrb. If autoantibodies not reacting with
Specificity of Autoantibodies 235

Rhnull RBCs were included, a total of 46 examples of


TABLE 7-3. “Rh” SPECIFICITY OF “WARM”
autoanti-Wrb were encountered. The incidence of
AUTOANTIBODIES DEFINED BY USE OF –D–, RHnull,
autoanti-Wrb in each group was 39.1% in AIHA; 12.1%
U–, AND Wr(b–) RED BLOOD CELLS (RBCs)
in methyldopa-induced positive DATs, and 26.7% in
normal blood donors with positive DATs. These inves-
The Only Plus Other
tigators23 commented that autoanti-Wrb can cause Autoantibody Autoantibodies
gross RBC destruction in vivo or can be benign on Specificity (% of patients) (% of patients)
occasions; it occurs with a higher frequency in AIHA
and “normal” donors with positive DATs than in “Simple” (Rh) 4.6 26
patients with methyldopa-induced positive DATs. We anti-nl* 4.6 23
anti-pdl* 3.4 33
would comment that if the group of AIHA with only anti-dl* 15 56
IgG on their RBCs (i.e., 38 patients) are analyzed, then anti-U 1.2 1.2
the percentage associated with anti-Wrb is lower, 28.9% anti-Wrb 2.3 37
compared with 39.1% of the total AIHA. If this group * Anti-nl, reacts with RBCs of common phenotype, but not –D– or Rhnull RBCs;
is compared with the methyldopa group (who also Anti-pdl, reacts with RBCs of common phenotype and –D–, but not Rhnull RBCs;
have only IgG on their RBCs), then the difference is not Anti-dl, reacts with RBCs of common phenotype, –D–, and Rhnull RBCs.
Modified from Issitt PD, Pavone BG, Goldfinger D, et al: Anti-Wrb and
as marked (i.e., 28.9% versus 12.1% instead of 39.1% other autoantibodies responsible for positive direct antiglobulin tests in
versus 12.1%); this difference is very similar to that 150 individuals. Br J Haematol 1976;34:5–18.
seen with the normal donors (70% of whom only had
IgG on their RBCs), compared with the methyldopa
group (i.e., 26.7% versus 12.1%) (Tables 7-3 and 7-4). with Wr(a+b–) RBCs, then anti-Wrb could be demon-
Issitt and coworkers23 also commented on strated in 41.8% of eluates, which is much closer to the
Worlledge’s report31 concerning autoanti-Ena and the 33% figure of “anti-Ena” described by Worlledge.31
weaker reactions seen with enzyme-treated RBCs. Issitt and coworkers23 believe that technical differ-
Issitt and coworkers23 had already shown that the Wrb ences may account for the fact that 73.6% of the 87
antigen was partially denatured by ficin, which would AIHA patients had anti-dl, while the incidence in 55
seem to support the concept that the antibodies patients studied by Vos and coworkers17 was 40%.
Worlledge reported might have been anti-Wrb. Bell and Zwicker33 confirmed that autoantibodies may
However, in tests on the unadsorbed eluates from 110 have anti-Wrb, anti-Ena, or a mixture of both
individuals with autoanti-dl, Issitt and coworkers23 specificities. Several other examples of autoanti-
only found four (3.6%) that failed to react with En(a–) Ena34-36 and autoanti-Wrb37,38 have been described.
RBCs, a very different incidence than the 33% Issitt and coworkers39 tested 119 sera, from patients
Worlledge reported.31 Issitt and coworkers23 thought with autoantibodies, with Di(b–) RBCs. None contained
that this difference may just reflect a difference in only anti-Dib but 2 of 74 sera subjected to adsorption
technique as they were able to show that in one case were shown to contain an autoanti-Dib component.
an ether eluate contained anti-Wrb + anti-dl, whereas Pr specificity is usually associated with cold autoan-
a heat eluate from the same RBCs contained only tibodies (see later) but is on rare occasions associated
anti-Wrb. When examples of anti-dl were adsorbed with WAIHA.36,40 Anti-Ena, -Pr, and -Wrb are often

TABLE 7-4. THE INCIDENCE OF ANTI-dl, “PURE” AUTOANTI-Wrb, AND AUTOANTI-Wrb AS A COMPONENT
OF ANTI-dl IN 150 PATIENTS WITH POSITIVE DIRECT ANTIGLOBULIN TESTS (DATs)

Percentage of
No. of Cases in Which
No. of Cases Tested anti-dl Was
No. of Cases Tested in Which Present in
Cases Tested in Which the anti-dl Which the
Total No. in Which anti-dl Present anti-dl Was, or
of Cases anti-dl Was Was Pure Contained, Contained,
Tested Present (%) anti-Wrb (%) anti-Wrb (%) anti-Wrb

AIHA 87 64 (74) 2 (2) 32 (37) 53


Aldomet-induced positive DATs* 33 23 (70) 0 4 (12) 17
“Normal” donors with positive DATs† 30 23 (77) 2 (7) 6 (20) 35

AIHA, acquired immune hemolytic anemia.


All eluates and sera containing anti-dl were absorbed with Wr(a+b–) RBCs to determine whether autoanti-Wrb was present. Anti-nl and anti-pdl did not
contain autoanti-Wrb because the D– and Rhnull cells used (one or both of which failed to react with these antibodies) were shown to be Wr(b+).
* The majority of these patients showed no evidence of increased rates of RBC destruction in vivo.
† As far as could be determined, none of these donors was undergoing an increased rate of RBC destruction in vivo.
Modified from Issitt PD, Pavone BG, Goldfinger D, et al: Anti-Wrb and other autoantibodies responsible for positive direct antiglobulin tests in 150
individuals. Br J Haematol 1976;34:5–18.
236 Immune Hemolytic Anemias

confused. For instance, when an antibody does not without effect. His DAT was initially negative but
react with enzyme-treated RBCs, anti-Ena, anti-Pr, and during his hospitalization it became weakly positive.
anti-Wrb should be among the list of suspects. If the During the next several weeks he was treated with
antibody also fails to react with En(a–) RBCs, which steroids, azothioprine, and blood transfusion without
lack glycophorin A, many workers call the antibody much success. During the fourth week of the disease,
anti-Ena. This can be a mistake, as anti-Pr and anti- he had a severe hemolytic transfusion reaction fol-
Wrb may also not react with En(a–) RBCs. The best lowing 500 mL of RBCs. During the fifth week of his
way of differentiating anti-Ena and anti-Pr is to test disease his RBCs were found to lack K, Kpa, and Jsa
the antibodies against neuraminidase (sialidase)- antigens, whereas the antithetical antigens k and Kpb
treated (NT) RBCs. As almost all sera contain anti-T, were only weakly expressed. His serum now con-
which will react with NT-RBCs, one has to use an tained anti-Kpb (titer of 256 by albumin technique).
eluate (prepared from RBCs incubated with the Following transfusion of Kp(b–) RBCs, the patient
serum in vitro) containing the antibody. Anti-Ena will began to improve (51Cr half-life of transfused RBCs
react with NT-RBCs and most anti-Pr will not. was 19 days). Serological tests performed 6 weeks
Unfortunately, one rare form of anti-Pr (anti-Pra) will after this first compatible Kp(b–) blood transfusion
react with NT-RBCs (see later); thus, the use of NT- revealed a very low level of anti-Kpb in the serum and
RBCs does not always differentiate anti-Ena from anti- a weakly positive DAT; anti-Kpb could be eluted from
Pr. En(a–) RBCs will adsorb anti-Pra but not anti-Ena. the cells. The patient’s RBCs now reacted strongly
Thus, to properly differentiate anti-Ena and anti-Pr with his serum collected 6 weeks previously, although
involves a great deal of work and access to relatively his RBCs were compatible with current serum, unless
large amounts of the rare En(a–) RBCs. Some anti-Ena the RBCs were ficin treated when they reacted
do not react with trypsin-, papain-, or ficin-treated weakly. The 51Cr half-life of the patient’s RBCs at this
RBCs (anti-EnaFS); some react with trypsin-, papain-, time was still short (8.5 days), with most of the RBCs
and ficin-treated RBCs (anti-EnaFR). As anti-Wrb does being destroyed in the spleen. During the eighth
not react with En(a–) RBCs, but does react with week of his illness, more Kp(b–) RBCs were trans-
trypsin-, papain-, and ficin-treated En(a+) RBCs, it can fused; although he had an excellent hematological
be confused with anti-EnaFR. The only way of differ- response, the 51Cr half-life of his own RBCs was only
entiating these two is to use RBCs from the only 8.5 days. The patient was discharged in excellent con-
example of the En(a+) Wr(b–) phenotype; anti-EnaFR dition after 4 months of hospitalization. By the 16th
will react with these RBCs but anti-Wrb will not. week, although the DAT was still weakly positive
Because of these difficulties, one has to read the liter- (complement only present on the RBCs), the anti-Kpb
ature very carefully when relying on specificities that in the serum was undetectable; the Kell phenotype
are said to be anti-Ena, -Pr, or -Wrb. was now normal, K– Kp(a–) Js(a–), with normal
WAIHA associated with M and N autoantibodies is expression of k and Kpb. After 7 months, the 51Cr
uncommon41-44; we described a fatal WAIHA associ- half-life of his own RBCs was 26 days. The authors
ated with an IgM autoanti-N.44 A few cases of anti-N suggested that although blocking of the Kpb antigen
associated with renal dialysis, and perhaps stimulated could have occurred during the acute phase of the
by formalin, were shown to cause shortened in vivo disease, the structure of the antibody molecule must
survival of N+ RBCs.45,46 No cases of AIHA due to have differed from that which normally has anti-Kpb
autoanti-M have been reported. Four cases of AIHA specificity because antiglobulin sera reacted with it
associated with autoanti-S have been reported.47-49 only weakly (i.e., very weak positive DAT). An alter-
In 1981 Reynolds50 described the first case of AIHA native and more probable explanation, they thought,
associated with autoanti-Ge. Several other examples was that during the acute stage of the disease, an
have been reported.51-55 Three of the four examples unknown exogenous factor, such as a virus, disturbed
presented with negative DATs, and their RBCs, the synthesis of Kell antigens by inhibiting the action
although typing as Ge+ did not react with the of transferases, which are necessary for the full
patient’s own anti-Ge at initial testing52-54; they are expression of these antigens on RBCs. Another very
discussed in the section below on autoantibodies similar case has been described.57
mimicking alloantibodies. In 1973 we described another association of anti-K
with WAIHA.58 The patient was a 49-year-old white
woman who presented in 1971 with intermittent
SPECIFICITIES ASSOCIATED fevers of unknown origin. She was found to have
WITH THE KELL SYSTEM granulomas of the lung and liver and to be anemic.
She was treated with tetracycline, isoniazid, and
As mentioned previously, there were two early pyridoxine. In December 1972, she was admitted to
reports of anti-K associated with WAIHA.12,13 In 1972, the hospital with fever and hemolytic anemia. Her
another case associated with Kell autoantibodies was DAT was found to be strongly positive. Her hemolytic
intensively studied in Warsaw and London.56 A anemia cleared spontaneously, and by January 1973
17-year-old boy developed severe hemolytic anemia she was hematologically normal and has remained so
and was treated with steroids and blood transfusion since. When we first studied her serologically at the
Specificity of Autoantibodies 237

end of 1972, her DAT was found to be strongly posi- adsorbed anti-Kx from an anti-KL serum much less
tive due to sensitization with IgG, IgM, and comple- efficiently than normal leukocytes. The patient’s DAT
ment. The eluate prepared from her RBCs contained became progressively weaker and, when last tested,
IgG and IgM anti-K. No reactions were obtained was barely positive. The anti-K had also diminished in
against K– RBCs. The patient’s serum contained IgG strength, as had the other antibodies reacting against
and IgM anti-K, reacting by IAT; enzyme-treated KK the K– RBCs. In contrast, the HLA-A2 and 7 titers had
RBCs were completely hemolyzed. The serum also not weakened. The ability of the serum to sensitize K–
contained another IgG antibody reacting by IAT, RBCs in vitro with anti-K appeared to diminish in
against 93% of 183 K– samples, including Ko, McLeod, direct relationship to the presence of RBC antibodies
Ch(a–), and Yk(a–) RBCs; positive reactions ranged in the serum.
from 1/2+ to 3+. The serum placed known weak, To summarize the above case: The patient was a
moderate, and strong Bg(a+) RBCs in order when female carrier of CGD who presented with hemolytic
tested blind and a tail of weak agglutination reactions anemia associated with RBC sensitization due to
was observed on titration (i.e., high titer, low avidity). anti-K. The patient was K– and we were able to
The patient’s serum also contained anti-HLA-A2, -B7, explain why the anti-K sensitized K– RBCs in vivo
and -A28, which have been shown to react with the Bg and in vitro. Although another antibody was present
blood groups on RBCs. The patient’s RBCs typed as in the serum, we were unable to demonstrate the pres-
AB K–k+ Kp(a–b+) Js(a–b+) Ku+ KL+. As Seyfried ence of this antibody together with anti-K in eluates
and coworkers56 had described an apparent depres- from K– RBCs, so we were unable to classify this as an
sion of Kell antigens associated with WAIHA, the example of the Matuhasi-Ogata phenomenon,27 but it
patient’s RBCs were tested by titration against several is of interest to note that Wilkinson and coworkers61
different anti-K, -k, and -Jsb reagents. No depression described in vitro adsorption of anti-D onto D– RBCs
of any Kell antigens was noted. Her lymphocytes in the presence of anti-Bg and that anti-Bg is highly
typed as HLA-A3, 11, 5, and 8. Her lymphocyte auto- suspect in our patient’s serum. Since reporting this
cytotoxicity was negative. When the patient’s serum case, Issitt and coworkers62 reported the phenomenon
was incubated at 4°C or 37°C with K– RBCs, it was of autoantibodies with mimicking specificities (see
possible to elute anti-K from these RBCs. The reaction later) and we now believe that the anti-“K” in this case
was always weaker than the eluate obtained from K+ was a “mimicking” antibody. The autoantibody was
RBCs. Eluates from these in vitro sensitized RBCs and probably not anti-K but only appeared to be and really
from the patients own RBCs were also tested for had a broader specificity. It appeared to be anti-K
anti-HLA activity with negative results. In 1971, because it reacted preferentially with K+ RBCs by the
Giblett and coworkers59 described a possible associa- AGT, but adsorption/elution studies showed that it
tion between the rare sex-linked chronic granuloma- was capable of reacting with K– and K+ cells.
tous disease (CGD) and the Kell system, in that there Viggiano and coworkers63 reported another mimick-
appeared to be a high incidence of the rare Ko pheno- ing anti-K in 1982. Several examples of autoanti-Kpb
type; the association was later found to be with the have been reported.56,57,64-66 Several examples were
McLeod phenotype (depressed Kell antigens) rather found in patients with weakened Kpb antigens56,57,64,65
than Ko.60 and are discussed in a later section on autoantibodies
As our patient presented with granulomas and an mimicking alloantibodies. Other high-incidence anti-
unusual serological phenomenon involving Kell, we gens in the Kell system have served as targets for RBC
investigated the family history carefully. We found autoantibodies.67-70 Marsh and coworkers67,68 tested
that the patient had one living healthy son of 26 years eluates from the RBCs of 950 DAT+ individuals against
of age, but another boy had died at the age of 9. His Ko RBCs, in addition to Rhnull, U–, and other RBCs.
case history and autopsy findings in retrospect were Four eluates contained autoantibodies to Kell system
classic for CGD, although not recognized as such at antigens; two were autoanti-Kpb, one was autoanti-
that time. The patient’s leukocytes were tested for K1367, and one was an autoantibody reacting with an
their ability to kill Staphylococci and Serratia. The undefined high-incidence antigen in the Kell system68;
killing effect was intermediate between leukocytes the latter patient had no evidence of HA. An example
from a normal individual and a patient with CGD. of autoanti-Jsb, enhanced by PEG, has been described71;
The leukocytes were also tested by the nitroblue tetra- this was not associated with WAIHA.
zolium dye reduction test; once again, the results
obtained were intermediate between normal and SPECIFICITIES ASSOCIATED WITH
CGD. Both sets of test results are diagnostic for a
carrier state of CGD. Marsh and coworkers60 showed THE KIDD AND DUFFY SYSTEMS
that anti-KL contained an antibody, anti-Kx, that
reacted with an antigen present on Ko RBCs. They Anti-Jka has been reported as an autoantibody on
further described a difference in the Kell antigens on several occasion,11,72-80 and three examples of
leukocytes from patients with CGD, in that they autoanti-Jkb have been reported.81-83 One of the exam-
appear to lack the Kx antigen. Dr. Marsh kindly tested ples of anti-Jka79 and one of anti-Jkb82 were found
the leukocytes from the patient and showed that they initially as mimicking antibodies (see later). It is of
238 Immune Hemolytic Anemias

interest to note that one anti-Jka was methyldopa associated with hypotension. Subsequently, clinical
induced,73 two were associated with Evans’s gout, obstructive lung disease, glomerulonephritis, and
syndrome,77,79 and one anti-Jka and two anti-Jkb were cirrhosis developed. An additional finding of
associated with bacterial infections.76,80,81 Two exam- extramedullary hematopoiesis indicated a long-stand-
ples of autoanti-Jk3 have been described82,84; both of ing hemolytic process. The authors speculated that
these were detected in pregnant women. Issitt and during this period, anti-A autoantibodies may have
coworkers85 tested 35 examples of anti-dl with combined with A antigen of tissue cells and that such a
Jk(a–b–) RBCs; none of the eluates contained anti-Jk3. phenomenon could have contributed to the multisys-
Four patients with autoanti-Fyb of the mimicking tem disease in a similar way to the humoral antibodies
type have been described86-88; the specificity was not that have been suggested for Goodpasture’s syndrome,
always clear as Fy(a+) but not Fy(a–b–) RBCs some- some cases of glomerulonephritis, fibrosing alveolitis,
times reacted.87 and chronic active hepatitis.
There are a few reports in the literature that describe
anti-A and anti-B in patients who have A and/or B
SPECIFICITIES ASSOCIATED antigens on their RBCs but these antibodies are not
WITH ABO AND Hh SYSTEMS true autoantibodies (even though the title of the article
sometimes uses this term). These are ABO antibodies
Although there are many reports in the literature of that developed following transplantation of kidney,
autoantibodies within the ABO and Hh systems, most liver, or hematopoietic stem cells. The antibodies
of these antibodies were “cold” autoantibodies of no appear to have been made in the A or B individuals by
clinical significance. There are several reports of donor lymphocytes that were infused with the donor
ABO autoantibodies being associated with hemolytic organ or donor stem cells. That is to say, “passenger
anemia, but most of these involve high thermal ampli- lymphocytes” from a group O donor proliferated in a
tude cold autoantibodies reacting optimally at 4°C group A recipient and produced allo-anti-A that would
(see later). There are only a few reports of anti-A appear to be an autoantibody (see Chapter 12).
or anti-B autoantibodies being associated with Kuipers and coworkers94 described an IgM mono-
WAIHA,11,89-93 and we find all these reports, except clonal (kappa) 37°C anti-H associated with malignant
reference 89, lacking in either clinical or serological lymphoma. The antibody reacted to a titer of 512 at
data to support the suggestion that the ABO autoanti- 37°C but no cold agglutinin titers were given; it is pos-
bodies caused WAIHA. For instance, there were few sible that this also was a cold autoantibody with a
data (e.g., indirect bilirubin, reticulocytes, lactic dehy- high thermal amplitude.
drogenase [LDH], haptoglobins, etc.) to support that a
hemolytic anemia was occurring in most of these MISCELLANEOUS TARGETS FOR 37°C
patients. It is also important to always exclude that the
patient had not received blood products (e.g., REACTIVE AUTOANTIBODIES
platelets, group O blood) containing ABO antibodies;
this was rarely mentioned in any of the publications. In 1972, we described a new target antigen for warm
A very unusual case of fatal fulminant intravascular autoantibodies, IT.95 Anti-IT had previously been
hemolysis associated with an anti-A autoantibody was described as a cold autoagglutinin of no clinical
described by Szymanski and coworkers.89 A group A1 significance.96 The “T” stood for “transitional” as it was
patient experienced severe bilateral lumbar pain associ- suggested that the putative antigen was a transitional
ated with generalized weakness, jaundice, and lethargy antigen appearing when i developed into I.96 If only
and was admitted the next day semicomatose. The cord (i) and adult (I) RBCs are tested, anti-IT may appear
patient was grossly icteric, had splenomegaly, and was to be anti-i as cord RBCs react much stronger than adult
severely oliguric. Eighteen hours later acute tachycardia RBCs, but in contrast to anti-i, adult i RBCs react very
developed, followed by respiratory and cardiac arrest. weakly (Tables 7-5 and 7-6). We first encountered an
On admission, the patient was found to have a strongly example of IgG autoanti-IT in a boy with Hodgkin’s
positive DAT (IgG but no complement). The patient’s disease but no hemolytic anemia.95 We later described
serum contained antibody(ies) that agglutinated all four cases of WAIHA associated with Hodgkin’s disease
RBCs tested (A1, A2, O adult, and O cord cells) at 22°C and IgG autoanti-IT.97 We questioned whether IT was
and reacted by IAT at 37°C; type O RBCs reacted much really an antigen transient between i and I, as we could
weaker. When the patient’s serum was diluted 1 in 5, A1 not demonstrate this by testing RBCs from fetuses at
and A2 RBCs still reacted (2+ at 4°C) and O RBCs were different gestation times.97,98 Hafleigh and coworkers99
negative; the reactions with group A RBCs could be found three examples of IgG autoanti-IT in patients who
inhibited by the addition of porcine A substance. A heat did not have Hodgkin’s disease or AIHA. Levine and
eluate prepared from the patient’s IgG sensitized RBCs coworkers100 studied 71 cases of Hodgkin’s disease;
demonstrated anti-A specificity by IAT at 37°C. It is seven were found to have positive DATs, and four of
interesting to note that the patient’s medical history was these were due to IgG autoanti-IT. Freedman and
noncontributory until 7 years before the onset of AIHA, coworkers101 described a case of AIHA associated with
when he was hospitalized for myocardial infarction an IgM anti-IT, reacting optimally at 37°C.
Specificity of Autoantibodies 239

body had an IgG component in addition to the IgM


TABLE 7-5. RELATIVE STRENGTH OF REACTIONS component. Sullivan and coworkers115 described a
SEEN WITH ANTI-I, ANTI-i, AND ANTI-IT case of WAIHA associated with autoanti-Kx; this is
still the only one in the literature.
I Adult i Cord i Adult
A “new” autoantibody specificity associated with
Anti-I 4+ 1+ 1⁄2+ AIHA was described by Marsh and coworkers116 in
Anti-i 1+ 3+ 4+ 1985. An IgG autoantibody, from a patient who died of
Anti-IT 1+ 4+ 1⁄2+ AIHA, reacted with all RBCs of common phenotype, Ko
RBCs, and RBCs treated with a dithiothreitol-papain
(ZZAP) solution. The antibody did not react with
Four examples of autoanti-Vel have been Rhnull RBCs or RBCs treated with 2-aminoethylisoth-
described.102-105 All of the antibodies were IgM. Two iouronium bromide (AET). As ZZAP and AET-treated
of the patients were DAT– and one had no evidence of RBCs exhibit loss of all Kell antigens, behaving like Ko
hemolytic anemia102; one patient was DAT– but had RBCs, the antibody did not appear to be reacting with
severe hemolytic anemia; and two patients were antigens belonging to the Kell system. The authors
DAT+, one had some evidence for increased RBC suggested that the antibody was reacting with an
destruction103 and the other had acute hemolytic antigen that is not part of the Kell or Rh system, but an
anemia.104 Three examples of autoanti-Sc1 and two antigen produced by in vitro membrane modification
examples of autoanti-Sc3 have been reported.106-108 by ZZAP, that is similar to antigens produced by in
The autoanti-Sc1 were associated with AIHA; the vivo membrane modifications known to occur on
autoanti-Sc3 were associated with positive DATs and Rhnull RBCs.116
anemia, but it was not clear from the data presented Table 7-7 lists the specificities that have been
whether AIHA was present. One example of autoanti- described as targets for “warm” autoantibodies.
Xga has been reported109; the anti-Xga was said to be
associated with hemolytic anemia; however, although
the patient was anemic, no data were presented to “MIMICKING ANTIBODIES”
support the diagnosis of hemolytic anemia.
Autoantibodies reacting with all RBCs except As early as 1956, cases were described where the
Lu(a–b–) cells have been reported.110-112 One example “wrong” (unexpected) antibody was eluted from the
appeared to be benign110 and the other was associated patients’ RBCs.117-119 Fudenberg and coworkers119
with hemolytic anemia.111 Marsh and coworkers110 described two cases where antibody eluted from the
showed that RBCs from Lu(a–b–) individuals of the patients’ RBCs had specificity for Rh blood factors not
recessive type would react with the antibody but present on the patients’ RBCs. Since that time many
Lu(a–b–) RBCs from individuals of the In(Lu) domi- workers have observed the same phenomenon, partic-
nant inhibitor type would not react, thus the antibody ularly anti-E being eluted from E– RBCs. In 1959,
was not directed against high-incidence Lutheran Dunsford and Stapleton120 reported on an anti-D that
antigens. The autoantibody was named anti-Wj by could be adsorbed and eluted from D– RBCs in vitro
Marsh and coworkers,110 but Wj appears to be the at 4°C; the authors suggested that the phenomenon
same as the previously described Anton (An) antigen, may be associated with the concomitant presence of a
so the autoantibody is now termed anti-AnWj. An cold autoagglutinin.
interesting association that is of great interest is that In a series of reports (1959–1964), Matuhasi121 and
the AnWj antigen has been shown to be a receptor for Ogata and Matuhasi122,123 showed that the “wrong”
Haemophilus influenzae.113 alloantibody can be adsorbed onto RBCs lacking its
In 1983 Denegri and coworkers114 described a case particular antigen if another specific antibody present
of WAIHA due to anti-Rx (initially called anti-Sdx). in the serum is adsorbed onto the cells. They showed
This antibody usually occurs as an IgM cold agglu- that if a mixture of anti-B and anti-D were present in a
tinin, but in the case described by Denegri114 the anti- serum and this serum was incubated with group B, D–

TABLE 7-6. ELUATE FROM RED BLOOD CELLS OF ACQUIRED IMMUNE HEMOLYTIC ANEMIA
PATIENT WITH SHOWING ANTI-IT SPECIFICITY

Dilutions of Eluate

Agglutination
Red Blood Cells 2 4 8 16 32 64 128 256 Score

I adult 1+ 1+ 1+ 1⁄2+ 0 0 0 0 18
i adult 2+ 1+ 1⁄2+ 0 0 0 0 0 12
i cord 3+ 3+ 2+ 1+ 1+ 1⁄2+ 0 0 32
240 Immune Hemolytic Anemias

elution process, concentration of any antibody present


TABLE 7-7. TARGET ANTIGENS FOR “WARM”
occurs, which will exaggerate the amount of antibody
AUTOANTIBODIES
adsorbed. Bove and coworkers126 did not explain why
this phenomenon occurs in only some patients.
Rh(“Rh”, c, C, D, e, E, f, rhi, G, Hr0, Rh34, Rh29, Rh39)
LW (LWa, LWab) In 1977 and 1978, Issitt and coworkers127,128 sug-
Glycophorin associated (U, M*, N, S, Ena, Pr, Wrb, Ge2, Ge3, gested that the “wrong” antibodies present in eluates
Ge4) may not have the simple specificity suggested by sero-
Kell (K, k, Kpb, K13, Jsb*) logical reactions against phenotyped RBCs but perhaps
Kidd (Jka, Jkb, Jk3)
Duffy (Fyb)
a less obvious, more complex, specificity. Auto-
Diego (Wrb, Dib) antibodies of seemingly “simple” Rh specificity (e.g.,
Scianna (Sc1, Sc3) anti-D, -C, -c, -E, rhi [-Ce], and -G), when tested with
ABO (A, B) RBCs having and not having the putative antigens,
Hh (H) including –D– and Rh null RBCs, by IAT, were studied
Others (Kx, Xga*, Co3, Ytab*, Vel, AnWj, IT, Rx)
by multiple adsorption tests. Seventy-one percent of
“Rh,” nonreactive with Rhnull red blood cells. these antibodies could be adsorbed using RBCs that
* Not associated, so far, with hemolytic anemia. lacked the specific antigen. Of interest was the finding
that if the antibody initially appeared to be autoanti-D,
there was a greater than 60% chance that it was a “true”
RBCs, then an eluate prepared from the group B cells autoanti-D (i.e., not adsorbed by D– RBCs). It was sug-
could be shown to contain anti-D as well as anti-B. In gested that those autoantibodies that at first appeared to
1967, Svardel and coworkers124 reported the adsorp- be directed against e, E, or c were only mimicking these
tion of anti-e, -f, and -Ce onto the RBCs of a cDE/cDE specificities and were probably anti-Hr or anti-Hro with
individual who had autoanti-I present in addition. a broad specificity. For instance, an autoantibody may
They suggested this was due to the Matuhasi- only react with E+ RBCs when a panel of RBCs is tested,
Ogata phenomenon. During the next decade, the but the reaction may be due to autoanti-Hr or -Hro,
“Matuhasi-Ogata phenomenon” became a popular mimicking anti-E, because E+ RBCs have more Hr/Hro
term to use when immunohematologists had no other than E– RBCs. This is comparable in some ways to a
explanation for a particular phenomenon, and in our weak (e.g., diluted) anti-A reacting with A1 and not A2
opinion, it has been greatly overused in the field by RBCs; the antibody is not anti-A1, but only reacting as
workers who usually had very little, if any, evidence such, because A1 RBCs have more antigenic sites than
to support their hypothesis. A2 RBCs, thus reacting better with weak anti-A.
One article appearing in 1969125 offered data to Similarly, a weak anti-LW can mimic anti-D because D+
suggest that the “Matuhasi-Ogata phenomenon” RBCs have more LW sites than D– RBCs. Issitt129
might be responsible for the adsorption of the termed the phenomenon “Matuhasi-Ogata maximus”
“wrong” antibody in autoimmune hemolytic anemia. and the phenomenon reported by Bove and cowork-
Later studies by Bove and coworkers126 seemed to ers126 as “Matuhasi-Ogata minimus.”
exclude this explanation, and indeed, one of the Issitt and coworkers127 found that sera of DAT+
authors (Issitt) of the 1969 publication125 has also patients may contain mimicking autoantibody alone
rejected his own earlier hypothesis.129 (e.g., autoanti-Hr reacting like anti-E), alloantibody
In 1977 Bove and coworkers126 criticized the sug- alone (e.g., anti-E) or mimicking autoantibody +
gested basis for the Matuhasi-Ogata phenomenon. alloantibody. They may be differentiated by adsorp-
These investigators used 125I-labeled IgG antibody tion; the mimicking antibody will be adsorbed by
and “nonantibody” IgG attached to RBCs and showed antigen-negative (e.g., E–) and antigen-positive (e.g.,
that there was no increased adsorption of antibody in E+) RBCs, whereas the alloantibody will only be
the presence of “nonantibody” IgG. They used a adsorbed by antigen-positive (e.g., E+) RBCs. It is
hypothetical case: If serum has 1.2 g% IgG and con- important to note that, in contrast to the earlier
tains 40 μg/mL of IgG anti-D, then 0.33% of the IgG is findings where the “wrong” antibody was eluted from
anti-D. Suppose 1 mL of D– RBCs are added and fol- DAT+ RBCs, of the 34 autoantibodies that had “mim-
lowing incubation the RBCs are well washed and an icking” specificities (i.e., adsorbed by “antigen-nega-
eluate prepared. Normal RBCs will bind 10 μg tive” RBCs) studied by Issitt and coworkers,127 28
IgG/mL of RBCs and eluates are known to contain (85%) were found in patients who had the putative
about 60% of the RBC-bound IgG. Thus, 1 mL eluate antigen present on their RBCs. In 150 patients with a
should contain about 6 mg IgG; 0.33% (0.02 μg) of this positive DAT and 87 with WAIHA, 4 (2.6%) and 43
IgG would be anti-D, which is enough to yield a pos- (49%), respectively, had autoantibodies with a true
itive antiglobulin test. In summary, Bove and cowork- “simple” specificity. Since this work with mimicking
ers126 thought that some IgG is always adsorbed by Rh antibodies, mimicking specificities in many
RBCs during incubation of RBCs with allogeneic systems have been described27,34,49-70,128-145 (Table 7-8).
serum. This is mainly “nonantibody” IgG but if In 1978,145 we published a report on an unusual
enough IgG is adsorbed, some of the molecules will be patient who not only had the “wrong” antibody
IgG antibody present in the serum and thus may be eluted from his RBCs but also was classified as a
present in an eluate made from the RBCs. During the “DAT-negative” AIHA (see Chapter 9). A previously
Specificity of Autoantibodies 241

TABLE 7-8. SPECIFICITIES ASSOCIATED TABLE 7-9. ANTIBODY TITERS (37°C IAT) OF
WITH MIMICKING ANTIBODIES SERUM FROM PATIENT WITH “MIMICKING” ANTI-E

System Antigens Red Blood Cells Tested

Rh D*, C*, E, c, e*, Ce, G, hrB* Serum rr R1R1 R2R2


MN S, Ena*
Lutheran Lub, “high incidence”* Unabsorbed 1 1 64
Kell K, Kpb*, “high incidence”* Absorbed with rr cells
Duffy Fya, Fyb* x1 0 0 8
Kidd Jka*, Jkb, Jk3 x2 0 0 2
Scianna Sc1*, Sc3* x3 0 0 0
Colton Coa Absorbed with R1R1 cells
LW LWa* x1 0 0 8
Gerbich Ge2, “Ge”* x2 0 0 2
Miscellaneous AnWj*, Lan x3 0 0 0
Absorbed with R2R2 cells
* Associated sometimes with depressed antigens (i.e., autoantibody mimicking x1 0 0 8
alloantibody).
x2 0 0 1
x3 0 0 0
untransfused 20-year-old man presented with a 7-day From Rand BP, Olson JD, Garratty G, et al: Coombs negative immune
history of malaise, fatigue, jaundice, dark urine, and hemolytic anemia with anti-E occurring in the red blood cell eluate of an
splenomegaly. Hemolytic anemia was indicated by a E-negative patient. Transfusion 1978;18:174–180.

hemoglobin of 8.7 g/dL, reticulocyte count of 8%,


bilirubin of 4.3 mg/dL (direct 0.1 mg/dL), and unde-
the enzyme reactive antibody had decreased to 16. Of
tectable haptoglobins. Tests for nonimmunological
particular significance is the decrease in the serum
mediated hemolytic anemia were negative. The DAT
anti–“E-like” antibody titer. In our experience, true
was repeatedly negative with polyspecific antihuman
alloantibodies usually are not affected during steroid
globulin, anti-IgG, -IgA, -IgM, and anti-C3. The
treatment although autoantibodies do decrease in
patient’s serum contained a weak anti-I, anti-E
titer. The results suggested that the serum anti–
strongly reactive by IAT, and an antibody reactive
“E-like” antibody in this patient behaved more like an
against all RBCs tested. The latter antibody reacted
autoantibody than an alloantibody.
weakly by IAT but strongly against enzyme-treated
Issitt and coworkers127,128 made some interesting
RBCs (titer 160). Eluates from the patient’s RBCs only
observations on this phenomenon. In 1977128 they
reacted with E+ RBCs. The patient typed E–. The
described a patient with similar serology to the patient
results of serum adsorptions with group O rr, R1R1,
above, except that their patient had a positive DAT.
and R2R2 RBCs are illustrated in Table 7-9. Following
They suggested that the anti-“E” might represent
the first and second adsorptions, the sera reacted only
autoanti-Hr rather than anti-E. They discuss that some
with the R2R2 RBCs and following three adsorptions
Rh alloantibodies are known to show marked prefer-
with either rr, R1R1, or R2R2 RBCs, there was no
ences for RBCs of certain phenotypes when IATs are
detectable antibody. These data indicate that the
performed but that adsorption studies reveal that some
serum antibody could be adsorbed to exhaustion by a
weakly, or even nonreactive RBCs carry small amounts
variety of Rh phenotypes although the serum reacted
of the antigens against which the antibodies are
only with E+ RBCs.
directed. For example, Rosenfield146 has observed that
The results of ether eluates prepared from the group
anti-hrS reacts better with RBCs from individuals who
O rr, R1R1, and R2R2 RBCs used for the adsorptions are
have ce cis genes than with those from individuals who
presented in Table 7-10. Regardless of the Rh pheno-
types used for adsorption, all eluates reacted only
with E+ RBCs. This was confirmed by repeated testing
with RBCs of varying Rh phenotypes. The possible TABLE 7-10. ANTIBODY TITERS (37°C IAT) OF
anti-c previously detected in the patient’s serum was ELUATES PREPARED FROM THE RED BLOOD CELLS
apparently too weak to be detected after the adsorp- USED FOR ABSORPTION IN TABLE 7–9
tion–elution procedures. Similar adsorption and
elution procedures were attempted with an eluate Red Blood Cells Tested
prepared from RBCs of the original patient specimen,
but the reactions were too weak to interpret with any Eluate from rr R1R1 R2R2
certainty. The patient was treated with high doses of
prednisone. By the twelfth day, his response allowed Absorbing rr cells 0 0 4*
Absorbing R1R1 cells 0 0 4*
the medication to be tapered, and 1 month from the Absorbing R2R2 cells 0 0 8*
onset of treatment, laboratory studies had returned
to normal. The DAT remained negative; however, * Anti-E specificity identified by larger panel.
From Rand BP, Olson JD, Garratty G, et al: Coombs negative immune
following recovery, anti-E could not be eluted from hemolytic anemia with anti-E occurring in the red blood cell eluate of an
the RBCs. Anti-E remained in his serum but the titer of E-negative patient. Transfusion 1978;18:174–180.
242 Immune Hemolytic Anemias

have Ce cis genes. In spite of this, almost all CDe/CDe to determine if the antibody (e.g., anti-E) is an alloanti-
bloods are hrS+. This means, of course, that in initial body or autoantibody, the best way is to perform
studies anti-hrS might well mimic anti-c or anti-f, but adsorptions with RBCs of different phenotypes (e.g., E–
that in adsorption studies CDe/CDe RBCs (which are and E+ RBCs). An alloanti-E should be adsorbed only by
c–, f–) invariably adsorb anti-hrS to exhaustion. A E+ RBCs, and by not E– RBCs, whereas, as discussed
similar situation exists with anti-hr,B which “prefers” previously, the autoanti-“E” will probably be adsorbed
RBCs from individuals with Ce cis genes to those from by E– as well as E+ RBCs.
individuals with ce cis genes. The original “Bastiaan” Autoantibodies can also mimic alloantibodies: Many
anti-hrB serum was believed to contain anti-C because patients have now been described who have depressed
of its preference for C+ RBCs, until it was found that this antigens at the time they have autoantibody in their
specificity could not be isolated by adsorption. In serum; sometimes the DAT is negative (but antibody
working with the sera from a series of individuals with can often be eluted from their RBCs). Thus, the auto-
highly exotic Rh phenotypes (Shabalala, Davis, antibody may appear like an alloantibody. Inves-
Santiago, Ellington, and Fentry), Rosenfield146 observed tigators have proved these are really autoantibodies by
some reactions that were very similar to those of the showing that when the hemolytic anemia is in remis-
“anti-E” in the eluate of the patient discussed in earlier sion and antibody is no longer detectable in the serum,
paragraphs. Although these observations have never the antigen strength returns to normal and the stored
been published in full, they were mentioned briefly in a serum, taken previously during the hemolytic episode,
1962 article by Rosenfield and coworkers.147 In several will now react with the patient’s RBCs. Some of the
of the sera studied, there were antibodies that showed alloantibodies mimicking autoantibodies referred to
very marked preferences for E or e. These antibodies above may also belong in this category because most
were shown not to be anti-E or anti-e by adsorption patients’ RBCs were not tested at different times for
studies with RBCs of appropriate phenotypes. It was antigen strength. Growing numbers of specificities
shown that an antibody with a preference for E could be have been associated with this phenomenon; these
readily adsorbed with E+ e–, or E+ e+ RBCs, but that E–, include Rh,127,128,130-133,145 LW,134,136 Kell,56-58,63,67,143
e+ samples would eventually totally adsorb the anti- Ge,51-54 AnWj,137 Duffy,138-140 Kidd,76,81,141 Ena,34 Co,142
body as well. In several instances the antibody makers Sc1 and Sc3,106,108 Lutheran,143,144 and Lan.144
were Hr–, so that their antibodies resembled the anti-Hr In 1996 Issitt and coworkers149 studied apparent
that is often made as an alloantibody by D– –/D– – or alloantibodies present in the sera of patients with
Dc–/Dc– individuals. Thus, Issitt and coworkers127,128 autoantibodies. Nineteen of the 117 (16%) sera appeared
feel that since autoanti-nl (i.e., autoantibody that reacts to contain obvious alloantibodies without further
with all RBCs of common phenotypes but not –D– or testing (e.g., adsorptions). Seventy-five (77%) of the sera
Rhnull phenotypes) reacts similarly to alloanti-Hr, then contained antibodies reacting with all RBCs tested.
the autoantibodies discussed above that appear to be Forty-one of these sera were adsorbed with autologous
anti-E may, in fact, be autoanti-Hr (anti-“Hr” or RBCs, revealing the presence of apparent alloantibodies
anti–“Hr-like”). We support this hypothesis. Data in 11 sera (27%); 69% of these were shown to be auto-
obtained from experiments on specificity of warm anti- antibodies mimicking alloantibodies (i.e., adsorbed
bodies using the AutoAnalyzer would also seem to by “antigen-negative” RBCs). When 34 sera were
support this concept.148 adsorbed with allogeneic RBCs, 14 (41%) appeared to
Issitt and coworkers128 also studied 48 autoantibodies contain alloantibodies; only 19% of these appeared to be
with apparent “simple” anti-Rh specificity (anti-e, -E, -c, mimicking antibodies. Thus, only 33% (not 41% as first
-D, -C, - Ce, -G) by means of multiple adsorption tests. suspected) of patients’ sera containing broadly reactive
They showed that 34 (70.8%) of these antibodies could autoantibodies appeared to contain true alloantibodies.
be adsorbed by RBCs lacking the antigens the antibod- It is interesting to note the differences between adsorp-
ies appeared to define. These autoantibodies that at first tions with autologous and allogeneic RBCs. Adsorption
appeared to be directed against e, E, or c antigens, most with allogeneic RBCs appeared to reveal more alloanti-
often had anti-Hr or anti-Hro specificity. Anti–“C-like” body specificity than adsorption with autologous RBCs.
autoantibodies may represent autoanti-Rh34, rather Also, apparent alloantibodies detected following
than anti-Hr or anti-Hro. Issitt and coworkers128 also adsorption with autologous RBCs were much more
comment that this new interpretation of the Rh-associ- likely to be autoantibodies mimicking alloantibodies
ated specificity does not change the philosophies (dis- than when sera were adsorbed with allogeneic RBCs.
cussed in Chapter 10) for the selection of blood for
transfusion. We would agree with this. If one believes
(as we do) that a patient with an autoantibody showing CHANGES IN SPECIFICITY
some autoanti-E specificity should receive E– blood, OF AUTOANTIBODIES
then it makes no difference if we now call this autoanti-
body autoanti-Hr, anti-“Hr,” anti-“E,” or anti–“E-like.” We (unpublished observations), and others150-152 have
As E– RBCs are reacting so much more weakly than E+ observed the specificity of warm autoantibodies to
RBCs (e.g., E– RBCs are sometimes negative by IAT), change during the course of a patient’s disease. In
then we would expect these cells to survive better in our experience it has usually been a broadening of
vivo than more strongly reacting E+ RBCs. If one wishes specificity. For instance, a patient will start with an
Specificity of Autoantibodies 243

autoantibody of “simple” Rh specificity and as the “mixed-field” appearance described by Stratton and
disease progresses the antibody will show more “non- coworkers154 and Ozer and Chaplin.156 We separated
specific” characteristics until it reacts with all cells “young” and “old” RBCs from fresh blood, by a capil-
tested, even by IAT. Other authors have described lary centrifugation method, and showed that the
other patterns. For instance, Beck and coworkers150 “stored RBC” antibody reacted preferentially with the
described a patient whose autoantibody over a 5-year older RBCs in fresh blood. This suggested to us that
period changed from “nonspecific” to specific; fur- the target antigen might be similar, or identical, to the
thermore, the specificity varied from anti-e to anti-c senescent cell antigen (SCA) described by Kay.159
and anti-f. The authors wisely comment on the fact Kay159 suggested that senescent cells are cleared from
that the treatment the patient was receiving (pred- the circulation because they develop an SCA that
nisone and methotrexate) may have affected the reacts with naturally occurring IgG autoantibodies to
specificity patterns seen. They suggest that particular SCA. These IgG-sensitized cells are then removed by
clones of immunocytes may have been selectively macrophages. Kay159 has characterized SCA biochem-
destroyed or suppressed. We have also seen patients ically; it is present on band 3 of the RBC membrane.
who first make a single alloantibody following trans- We were able to demonstrate that the RBC autoanti-
fusion, then make more alloantibodies, then eventu- body that reacted preferentially with stored and “old”
ally make autoantibodies (see Chapter 9). RBCs could be inhibited in vitro with synthetic SCA
that was kindly provided by Dr. Kay.159 Thus, we
believe that “stored” RBC antibodies are reacting
SPECIFICITIES NOT ASSOCIATED against SCA and that the ubiquitous anti-SCA, which
WITH BLOOD GROUP ANTIGENS is usually present in very low levels, like the ubiqui-
tous anti-I, can become pathogenic on occasions.
There are an increasing number of observations relat- Gray and coworkers160 found that reticulocytes
ing to causes for positive DATs and reactions of have 60% of the D content of mature RBCs. They also
serum antibodies with all, or most, RBCs including showed that IgG autoantibodies eluted from the
the patient’s own RBCs, that are not associated with RBCs of patients with AIHA reacted weaker with
blood group antibodies. Table 7-11 lists these with reticulocytes than mature RBCs in four of five cases.
appropriate references. Branch and coworkers161 performed DATs on age-
fractionated RBCs of 24 DAT+ patients. Seventy-nine
Optimal Reactions with Stored percent of the DATs were stronger when the older
or “Old” RBCs RBC fraction was tested (type 1); 37% of the reticulo-
cyte-enriched fractions had negative DATs. Twenty-
Since 1952, there have been reports in the literature of one percent of the autoantibodies seemed to show no
antibodies reacting better with stored RBCs than fresh preference for old or young RBCs (type II). Branch
RBCs.153-158 These antibodies usually were cold and coworkers161 suggested that type I warm
autoagglutinins (see later), but sometimes reacted up autoantibodies might be recognizing an as yet
to 37°C. Some of the patients having such antibodies unidentified RBC antigen, possibly a cryptantigen
had hemolytic anemia, but it was uncertain whether closely associated with the Rh peptide but not fully
this was caused by the cold autoantibody reacting expressed on very young RBCs. They hypothesized
optimally with stored RBCs. that the type I autoantibody might represent aug-
In 1989, we reported a patient with WAIHA associ- mented production of the physiological autoanti-
ated with an autoantibody that reacted preferentially body responsible for clearing senescent RBCs. We
with stored RBCs.158 The antibody showed similar would suggest that the antibody against stored RBCs
characteristics to other antibodies described previ- (and older RBCs), that we showed was anti-SCA,158
ously in that it reacted well with enzyme- and heat- is identical to some of the type I autoantibodies161
treated RBCs. It also showed the characteristic and that these are indeed directed against SCA.
Finally, Herron and coworkers162 described an e+
patient with AIHA associated with a negative DAT
but a strongly reacting IgM anti-e. Anti-e was eluted
TABLE 7-11. THE 37°C REACTIVE ANTIBODIES from the patient’s RBCs. The anti-e reacted much
THAT REACT WITH ALL OR MOST RED BLOOD weaker with the patient’s RBCs than other e+ RBCs in
CELLS (RBCs), INCLUDING PATIENT’S OWN RBCs vitro. The anti-e reacted preferentially with “older”
(IN VITRO AND IN VIVO) BUT ARE NOT DIRECTED RBCs when age-fractionated RBCs were tested. When
AT BLOOD GROUP ANTIGENS the patient went into remission, the patient’s RBCs
reacted equally strongly as other e+ RBCs, with the
Antibody to References
patient’s stored serum from the time of the hemolytic
Older or stored RBCs 153–162 episode. The authors suggested that the patient’s
Younger RBCs 34, 163–166 RBCs that survived destruction during the hemolytic
RBC membrane proteins 167–173 episode were younger RBCs; they had less e antigen
Phospholipids 174–180 and thus, very little antibody on them, yielding a neg-
Ig idiotypes 181, 182
ative DAT but a reactive eluate.
244 Immune Hemolytic Anemias

strongly suggest that aplastic crises in some patients


Optimal Reactions with Younger RBCs with AIHA can be due to autoantibodies directed
When Branch and coworkers161 performed DATs on against targets on erythroid progenitor cells. Davidyuk
age-fractionated RBCs from DAT+ patients, they found and coworkers166a reported that four of five patients
that none of the 24 autoantibodies they tested reacted with anti-phospholipid antibody syndrome had auto-
better with younger (reticulocyte-rich fraction) RBCs antibodies to immature RBCs. These antibodies inhib-
than older RBCs. Nevertheless, there is some evidence ited the growth of hematopoietic precursor cells in vitro.
that RBC autoantibodies sometimes react preferentially
with targets on younger RBCs. Hedge and coworkers163
reported on three patients with WAIHA who had low Miscellaneous Membrane Components
reticulocyte counts when they were most anemic and in as Targets for RBC-Bound
whom no RBC autoantibodies could be detected by the Autoantibodies
DAT. They postulated that reticulocytes may be selec-
tively destroyed if antibodies are directed against IgG autoantibodies to RBC skeletal proteins have also
targets on young RBCs, thus giving rise to a population been detected in human sera. Lutz and Wipf167 detected
of cells whose target antigenic sites are poorly expressed autoantibodies to spectrin, actin, and band 6 in the sera
(i.e., older RBCs). Conley and coworkers164 reported on from all 10 healthy donors that were tested. As these
five cases of WAIHA with reticulocytopenia; all five had proteins are not present on the surface of the RBC mem-
rapid-onset life-threatening anemia. Repeated aspira- brane, one would think that RBC-bound IgG autoanti-
tion and biopsy of the marrow revealed that it was bodies to such proteins would not be detected on
hyperplastic and packed with erythroid cells showing a normal RBCs. However, Wiener and coworkers168
normal pattern of differentiation. Polychromatic RBCs eluted IgG antispectrin from the RBCs of patients with
were seen in marrow smears but none were observed in β-thalassemia but not from the RBCs of patients with
peripheral blood smears. Unlike the cases described by sickle cell anemia or normal donors. They suggested
Hedge and coworkers163 the DATs in all five cases were that this IgG may play a role in the increased rate of
positive and autoantibodies were present in the sera. destruction of RBCs in thalassemic patients. Wakui and
The eluate and serum of one of the patients were tested coworkers169 recently described an autoantibody to
against reticulocyte-rich and reticulocyte-reduced RBCs. RBC protein 4.1 in a patient with AIHA. This antibody
The autoantibody reacted weaker against the reticulo- was detected, together with “anti–Ena-like” and anti-S,
cyte-reduced fraction than the reticulocyte-rich fraction. in the patient’s serum. It was unclear whether the anti-
Reticulocyte-reduced RBCs sensitized with autoanti- S was an autoantibody or alloantibody. Unfortunately,
body reacted better in an in vitro monocyte phagocytic an eluate from the patient’s RBCs was not tested for
assay than reticulocyte-rich sensitized RBCs. Thus, anti-protein 4.1 activity. An eluate prepared from S+
there seemed no evidence for an antibody directed RBCs incubated with the patient’s serum did not
against a target present mainly on reticulocytes. contain antiprotein 4.1.
In 1983 Garratty and coworkers34 described a case of In the section on mimicking antibodies, it was men-
pure red cell aplasia associated with a mimicking anti- tioned that antigens are sometimes depressed in AIHA
Ena. It was suggested that the autoanti-Ena had when the autoantibody is first detected. This depres-
destroyed a population of young RBCs in the marrow. sion can be severe enough that the RBCs do not react
Hauke and coworkers165 described a patient with with typing sera. Sometimes the patients have been
severe WAIHA, reticulocytopenia, RBC hyperplasia of shown to have RBC membrane abnormalities. Garratty
the bone marrow, and a positive DAT (IgG and comple- and coworkers34 described a case of AIHA and red cell
ment). The patient was found to have an autoantibody aplasia associated with a “mimicking” autoanti-Ena
to early erythroid progenitors (BFU-E). It was unclear where the patients RBCs reacted with some, but not all,
whether this was in addition to the antibody reacting anti-Ena. Examination of the RBC membrane by
with mature RBCs. Mangan and coworkers166 described sodium dodecyl sulfate–polyacrylamide gel elec-
a similar case but provided much more data. The trophoresis (SDS-PAGE) revealed only a slight reduc-
patient had first presented with AIHA and a 60% retic- tion of glycophorin A. The RBCs from an En(a+)
ulocyte count. The IgG autoantibody on the RBCs was patient with an anti-EnaTS, which caused a fatal
identified as anti-e. The patient responded to steroids. hemolytic transfusion reaction, showed greater abnor-
Two years later the patient presented again with severe malities of the membrane.170 The RBC membranes
anemia. The autoantibody now reacted with all RBCs were shown to contain glycophorins B and C by SDS-
tested except Rhnull, although an anti-e component PAGE with periodic acid–Schiff’s base staining with
could be demonstrated. The reticulocyte count ranged weak staining of components in the regions correspon-
from 0.4% to 0.6%, and erythroid precursors were virtu- ding to monomers and dimers of glycophorin A and
ally absent from the bone marrow. The serum was heterologous dimers of glycophorins A and B. The
found to contain a complement-dependent IgG nature of these components was not identified, but
inhibitor directed against CFU-E and BFU-E. The ery- their presence suggested that the patient’s RBCs
throid progenitor cell inhibitor was still present follow- expressed a previously undescribed glycophorin A
ing adsorption of the serum with e+ RBCs, and was not variant. Several patients have been described with
present in an eluate from the patient’s RBCs. These data abnormalities of glycophorin. Beattie and Sigmund51
Specificity of Autoantibodies 245

reported a case of AIHA and aplasia, associated with described a case of AIHA associated with high titer IgM
autoanti-Ge. The RBCs of this patient showed an cardiolipin antibodies. An interesting observation was
altered glycophorin C. Reid and coworkers171 studied that the cardiolipin antibodies bound more efficiently to
the immunochemical specificity of the target antigens stored RBCs than fresh RBCs; bromelin-treated RBCs
reacting with autoanti-Ge from two patients with reacted better than stored untreated RBCs. In contrast,
AIHA. One anti-Ge reacted with normal, but not with Davidyuk and coworkers166a found that four of five
abnormal glycophorin C, associated with Ge– RBCs. autoantibodies from patients with anti-phospholipid
This patient’s RBCs had an alteration of glycophorin C antibody syndrome reacted better with immature RBCs.
with the other glycophorins being normal. The
autoanti-Ge from the other patient was similar to
alloanti-Ge3 in that it reacted with both glycophorins C Idiotypic Targets on RBC-Bound
and D from normal RBC membranes and with the IgG Autoantibodies
abnormal glycophorin C found in RBCs from indivi-
Masouredis and coworkers181 reported that eluates
duals with Ge– RBCs of the Yus type.
from DAT+ donors contained IgG antibodies to idio-
There have been other isolated reports of RBC mem-
typic determinants (anti-id) on the RBC-bound IgG
brane abnormalities associated with AIHA but these
autoantibody, in addition to RBC autoantibodies (see
have not related to specific autoantibody specificity.
Figure 9-2 on page 321). Eight of 12 eluates prepared
Gomperts and coworkers172 studied RBCs from nine
from the RBCs of DAT+ donors contained at least two
cases of AIHA. RBC stroma was extracted using dilute
populations of IgG autoantibodies. The first population
acetic acid and the isolated protein electrophoresed on
reacted with RBCs by the IAT; this was the RBC autoan-
8 M urea starch gel. Six of the nine cases showed the
tibody. The second population did not react with
absence of one band and absence or decreased intensity
untreated RBCs but would directly agglutinate IgG
of a second band. The pattern was the same as that
(anti-D)-coated RBCs; this was the anti-id. In a later
observed when RBCs from hereditary spherocytosis
article, eluates from the RBCs of two patients with
were tested. The eluates from RBCs of the six cases with
AIHA were shown to contain RBC autoantibody but no
absent bands all showed definable “specificity” (e.g.,
anti-id.182 The antibodies were designated as anti-id by
anti-nl, -pdl, etc) but the remaining three were
classified as “nonspecific.” Kajii and coworkers173 iso- Masouredis and coworkers182 because the autoanti-
body appeared to be directed against determinants on
lated a 12-kDa peptide from the RBCs of four patients
the Fab portion of IgG (only one antibody was studied),
with AIHA. This peptide could not be obtained from
and the antibody would agglutinate RBCs coated with
RBCs of 50 normal persons, or patients without AIHA.
anti-D, but not antibodies of other specificities.
In one patient, with angioimmunoblastic lympha-
denopathy and dysproteinemia (AILD), the peptide
In 1999, Garratty reviewed specificities that have
was still detectable following treatment, when the RBC
been reported to be associated with autoantibodies
autoantibodies had disappeared, but was no longer
reacting optimally at 37°C.183
detectable when the patient’s AILD was in complete
remission, following chemotherapy.
SPECIFICITIES ASSOCIATED WITH
Antibodies to Phospholipids COLD AGGLUTININ SYNDROME
Antibodies to phospholipid (e.g., anticardiolipin) may
attach to RBC membranes, leading to a positive Nonspecific cold autoagglutinins were first reported
DAT174,175 and possibly AIHA.166a,176-180 Hazeltine and by Mino in 1924.184 The story of the unraveling of
coworkers174 detected cardiolipin antibodies in 22% of their specificity closely parallels the history of the Ii
patients with systemic lupus erythematosus (SLE). The blood group antigens.
presence of such antibodies in the serum was associated Three main groups of antigens recognized by
with a positive DAT. Analysis of eluates from the human cold agglutinins (CAs) have been defined on a
patient’s RBCs and adsorption studies showed that serological and biochemical basis. The first group
some cardiolipin antibodies may react with phospho- consists of the Ii antigens. They are protease- and
lipid on RBC membranes. Win and coworkers175 sialidase-resistant differentiation antigens. I antigen
reported on positive DATs, associated with phospho- is fully expressed on adult and i antigen is fully
lipid antibodies, in blood donors. Sthoeger and cowork- expressed on fetal RBCs. Anti-i recognizes linear poly-
ers176 showed the presence of cardiolipin antibodies in N-acetyllactosamine or type 2 chains, which are con-
the sera and eluates from RBCs of two SLE patients with verted into I antigens in the first year after birth by
AIHA. Following steroid treatment one patient went branching. The second group consists of the Pr and Sa
into remission, the DAT and IAT became negative, and antigens. They are not differentiation antigens on
cardiolipin antibodies were no longer detectable. The RBCs but are expressed in equal strength on adult and
other patient remained DAT+, and cardiolipin antibod- newborn RBCs. Pr1,2,3 antigens are destroyed by pro-
ies were still detectable. The authors suggested that the tease and sialidase treatment of RBCs, whereas Pra is
cardiolipin antibodies may play a direct role in the inactivated only by proteases. Anti-Pr and anti-Sa
pathogenesis of the AIHA. Cabral and coworkers177 recognize the sialo-O-glycans of glycophorins. The
246 Immune Hemolytic Anemias

third group consists of the Sia-11, Sia-bl, and Sia-1b1 anemia. Later in the same year, Marsh and Jenkins187
(formerly termed Vo, Fl, and Gd) antigens. They are and, in 1961, Marsh188 described the first two exam-
susceptible to sialidase but are resistant to proteases ples of anti-i; antibodies that appeared to react anti-
on RBCs. Sia-1l and Sia-b1 antigens are differentiation thetically to anti-I. Using anti-I and anti-i, Marsh was
antigens created by sialyation of linear and branched able to show that unlike other blood group systems so
type 2 chains, respectively. Sia-1l and Sia-b1 resemble far described, cord RBCs from newborn babies were
i and I antigens. Another antigen of this group, rich in i antigen and possessed very little I antigen.
Sia-1bl, is not developmentally regulated, but is The I antigen slowly developed at the expense of i
expressed in equal strength on adult and newborn until at least 18 months of age, at which time adult
RBCs. It is detected by anti–Sia-1bl CAs recognizing status was reached. Adult RBCs normally are rich in I
linear as well as branched sialylated type 2 chains. antigen but have only small amounts of i antigen
present. Some adults are found to give intermediate
reactions, IINT (i.e., weaker I antigen). Rare adults are
Ii Blood Group Antigens and Antibodies found whose RBCs have less I antigen and more i
In 1956, Weiner and coworkers185 studied an unusual antigen than cord RBCs; these are the i adults.
patient with cold agglutinin syndrome (CAS) who Marsh188 suggested the following order of increasing
had severe transfusion reactions, even if the donor strength of I: i1, i2, icord, IINT, I; i1 is usually associated
blood was kept warm. They were energetic enough to with white individuals, and i2 is usually associated
screen RBCs from over 22,000 donors with the with black individuals.
patient’s serum, finding 5 to be compatible (4 blacks, Although cord RBCs react much weaker than adult
1 white). No correlation was found with the Lewis, RBCs at all temperatures when tested with a low titer
Lutheran, Duffy, Kidd, P, Vel, or ABO systems. The cold agglutinin, the results can be very confusing
authors concluded they were dealing with a new when a pathological high titer cold agglutinin is
blood group specificity, which they designated I (to tested; very little difference in titer is noticed at 4°C,
indicate its high degree of individuality). The antibody but the specificity becomes more obvious as the tem-
was thus anti-I, and the rare individuals whose RBCs perature is raised. Table 7-12 shows typical reactions
were compatible with the serum (containing anti-I) of adult and cord RBCs with a pathological high titer
were designated i, or “I-negative.” Although RBCs anti-I. It should also be noted that various examples of
from the five donors did not react at room tempera- cord RBCs can show marked differences in reactivity
ture when untreated, they all reacted at 4°C and at with some anti-I.
room temperature when ficin-treated. A feature of In 1970,190 a powerful anti-I was described that was
Weiner’s case that we find hard to understand is that strongly inhibited by hydatid cyst fluid and inhibited
the patient’s own washed RBCs were not agglutinated to a varying extent by all of the 181 human saliva
by his own serum at room temperature, unless the samples tested; previously, saliva had been reported
RBCs were enzyme-treated; in fact, they reacted simi- not to inhibit anti-I.191 Infants at birth and i adults
larly to the so-called i donors! were also found to have high concentrations of I sub-
In 1960 Jenkins and coworkers186 found that 50 sera stance in their saliva. This investigation suggested
containing weak cold autoagglutinins that had previ- that I antibodies are of two kinds—those inhibitable
ously been called “nonspecific cold agglutinins” had by saliva and those not inhibited; this confirmed an
anti-I specificity; none of these donors had hemolytic earlier suggestion by Marsh who had termed the two

TABLE 7-12. REACTIONS OF A TYPICAL ANTI-I ASSOCIATED


WITH COLD AGGLUTININ SYNDROME

4°C 25°C 30°C

Dilution of Patient’s Adult Cord Adult Cord Adult Cord

2 4+ 4+ 4+ 1+ 1+ 0
4 4+ 4+ 4+ 1+ 1⁄2+ 0
8 4+ 4+ 3+ 1⁄2+ 0 0
16 4+ 4+ 3+ 0 0 0
32 4+ 4+ 3+ 0 0 0
64 4+ 4+ 2+ 0 0 0
128 4+ 3+ 1+ 0 0 0
256 3+ 3+ 1⁄2+ 0 0 0
512 3+ 2+ 0 0 0 0
1024 2+ 1+ 0 0 0 0
2048 1+ 0 0 0 0 0
4096 0 0 0 0 0 0
Specificity of Autoantibodies 247

types Ia and Ib. Marsh and coworkers191 found that reacting optimally at 0°C to 4°C; and almost always
human milk contained a high concentration of water- reacting up to 30°C to 32°C and rarely reacting at
soluble I blood group substance. Tests with 24 differ- 37°C, when saline-suspended RBCs are used (see
ent anti-I showed that, to a variable extent, all of them Table 7-12). It is by far the most common antibody to
could be inhibited by milk and some could be inhib- be associated with CAS (both the chronic idiopathic
ited by strong I secretor saliva. This susceptibility to cases and those secondary to Mycoplasma pneumoniae
inhibition was not related to titer, and the results sug- infection). In our own series, 49 of 54 cases (91%) were
gested that qualitative differences in the antibody associated with anti-I. Other workers have reported
antigen reactions was responsible. In 1971, Marsh and similar findings. Anti-i is not commonly detected in
coworkers189 described two components of the I normal serum as is anti-I but has been reported to be
antigen, which they named IF (fetal) and ID (devel- present in as high as 70% of sera from patients with
oped). The IF component was found to be present on infectious mononucleosis. When it is detected in other
all human RBCs, including those of icord, iadult, and cases, it is often associated with diseases of the reticu-
also on rhesus monkey RBCs. The ID component loendothelial system. When it is present in high
develops slowly on the RBCs before birth and, to a enough titer and its reactivity is of high thermal range,
greater extent, 18 months after birth. Inhibition it can cause AIHA. Of 54 cases of CAS in our series,
studies with human milk showed that strongly 4 (7.4%) were associated with anti-i. Although the
inhibitable anti-I were of the anti-ID variety, but only a clinical histories are very limited, there is a suggestion
minority of such sera were inhibitable. Naturally that one of the first two patients to be described as
occurring low titer cold autoagglutinins were found having anti-i had AIHA.187 This patient came under
mainly to be anti-ID, whereas the high titer cold investigation for suspected reticulosis and moderate
autoagglutinins associated with AIHA were found anemia. An anti-i reacting to a titer of 256,000 with
commonly to contain anti-IF, either alone or together iadult RBCs was present in his serum, reacting strongly
with anti-ID; rarely, anti-ID was encountered exclu- at room temperature and weakly at 37°C. The anti-i
sively. This suggested an explanation for the strong only reacted to a titer of 4 to 16 with Iadult RBCs. Often,
reaction of high titer anti-I associated with AIHA, even strong anti-i (i.e., high titers against iadult and
with cord RBCs, which are rich in IF but have very icord RBCs) will not react or only react weakly at room
little, if any, ID. Marsh and coworkers189 also dis- temperature with normal adult RBCs, including the
cussed the possible place of IF and ID in the develop- patient’s own. Thus, hemolytic anemia is rare even
ment of the I antigen. when a powerful anti-i is present. Sometimes the i
Dzierzkowa-Borodej and coworkers192 suggested antigenic status of the patient’s RBCs can be
that the anti-I that are inhibitable are different than increased,193,194 thus making the patient’s RBCs more
anti-ID. Although they act similarly serologically, they reactive with anti-i than normal adult RBCs. When
can be differentiated by their capability to precipitate performing cold agglutinin titers and thermal ranges,
in gel at 4°C. Saliva, colostral IgA, and desialized gly- etc., it is always useful to include tests against the
coprotein from RBCs gave strong precipitin lines with patient’s own RBCs.
inhibitable anti-I but not with other anti-I. They sug- Since the original description of anti-i, other cases of
gested that the inhibitable anti-I be called anti-IS. In an CAS caused by anti-i have been described.195-198
extensive study on 12 anti-I cold autoantibodies (one Chemistry of Ii Antigens. Ii antigens are closely
of normal titer 32, the others ranging from 256 to 2 × related to ABO, H, and Lewis blood group antigens. A
106), they found that only 4 of 12 sera demonstrated number of antibodies have been described that will
single specificity (1 anti-IS, 3 anti-IF) and that the only react when I is present together with other antigens
others demonstrated two or more specificities from the ABO, Hh, and Lewis systems (e.g., anti-IA,
(1 anti-ID + anti-IS, 1 anti-ID + anti-IF, 3 anti-ID + anti-i, -IH, -iH, -IB, -IP, -IP1, -iP1, -ITP1, and anti-LebH). These
and 2 anti-ID + anti-IF + anti-IS). These differences antibodies appear to react with complex antigens dis-
could not be easily determined by using Iadult, icord, tinct from the separate antigens, for example, anti-IB
and iadult RBCs but became more obvious when the will only react with RBCs containing both I and B anti-
RBCs from a rare adult having only IF were used gens, and not with RBCs containing I but not B or B but
(these RBCs failed to react with anti-ID and anti-i but not I; the amount of IB on RBCs is not related to the
were agglutinated by anti-IF). It is interesting to note amount of I or B on the RBCs,199,200 thus appearing to be
that the same authors have shown that desialization a completely separate “antigen.” In 1971 Feizi and
of RBCs enhanced their agglutinability by anti-ID and coworkers201 analyzed the I-active antigen extracted
anti-IS but had no effect on their reaction with from human milk. The sugar composition of this mate-
anti-IF.193 rial closely resembled the chemistry associated with
It has been clear over the years that autoanti-I can ABO blood group substances; however, its content of
exist in two forms: (1) a low titer autoantibody occur- frucose was unusually low. In this respect, it resembled
ring in almost all normal human serum as a naturally blood group precursor-like substances that had been
occurring cold agglutinin; reacting optimally at 0°C to isolated from human ovarian cyst fluid lacking ABO
4°C; rarely acting above room temperature; and (2) a and Lewis substances.202,203 Feizi and coworkers201,204
high titer autoantibody often associated with CAS; suggested that I specificity was concealed in interior
248 Immune Hemolytic Anemias

structures of the blood A, B, H, Lea, and Leb substances could be recovered form precipitates prepared with
and may be exposed by stepwise periodate oxidation anti-A and -B precipitating sera. Thus, the water phase
and Smith degradation of ABO and H substances. Thus, left after extraction of stroma with n-butanol com-
I substance would seem to be a precursor for the biosyn- prises a single antigenic material in which all A, B, H,
thesis of H, A, and B. In a later study, the same workers and I blood group activities are located on the same
studied 11 anti-I sera and found they could be divided molecule. This conclusion offers an explanation for
into at least six groups based on their reactions with the anti-I that are influenced by ABH group of the test
human milk, ovarian cyst fractions (containing “ABH” RBCs (e.g., anti-IH, -IA, -IB) in that the adjacent A, B,
precursor blood group substances), degraded ABH sub- H, and I active structures may give rise to antibodies
stances, and hydatid cyst fluid. Four of five anti-i resem- of mixed ABH and I specificity. The results obtained in
bled each other, but the fifth differed in its reaction with this study would agree with Feizi and coworkers201
hydatid cyst fluid and milk. and Marcus and coworkers209 that terminal galac-
Burnie195 and Cooper and coworkers205 have topyranosyl residues are part of the I structure. It is
shown that normal plasma can be shown to contain I interesting to note that the isolated I and H blood
and i substances in small quantities. Rosse and Lauf206 group substances were easily adsorbed onto human
extracted the antigens (“I”) reacting with cold autoag- RBCs in vitro and thus group Oi RBCs could easily be
glutinins from human RBCs, using n-butanol. A fasci- changed to OI RBCs. One finding contrasted with the
nating observation was made that although the finding of Rosse and Lauf206 described earlier; iso-
antigen and antibody were not able to react at 37°C lated preparations of I active antigens precipitated
when the antigen was present in the intact RBC after only at 4°C and no reactions occurred at 37°C.
solubilization, antigen and antibody reacted equally Red cell glycoproteins contain two kinds of
well at 37°C and 0°C. In addition, the amount of “I” oligosaccharide chains: (1) alkali-labile chains consist-
antigen extracted from the RBCs of adult and new- ing of galactose, N-acetygalactosamine, and a large
borns appeared to be about the same! In a previous proportion of sialic acid; and (2) alkali-stable chains
study,207 it had been suggested that the effect of tem- containing galactose, N-acetyglucosamine, mannose,
perature changes was not on the antibody but was and low amounts of sialic acid and fucose.210-214
rather on the antigen or the RBC membrane. This con- Aklali-stable chains are required for I activity, whereas
clusion was based on the fact that the change in MN activity depends on alkali-labile chains. Red cells
affinity of antibody for RBCs of newborn infants (icord) contain one major glycoprotein (glycophorin) and a
or of iadult RBCs was very much greater than that for few minor ones. I activity has been reported to be
RBCs of normal adults. Because any effect on the anti- associated with the minor glycoprotein by some
body of change of temperature would have been the workers215 and glycophorin by others.216 Lisowska
same in both situations, they concluded that a change and coworkers193 tested three fractions of red cell gly-
in configuration of the antigen or the RBC surface coproteins obtained from Sepharose 4-B chromatogra-
must be responsible for the characteristic cold reactiv- phy for I activity with 10 anti-I sera. Fraction 1 was
ity of the antigen–antibody interaction. They further further purified by separating ABHI-active substances
concluded that their later experiments indicated that from MN active sialoglycoprotein. This fraction had
the reactivity of cold agglutinins only in the cold was the greatest I activity and contained the lowest
due to an effect of cold on the RBC membrane, amount of alkali-labile obligosaccharide chains. The
because the antigen–antibody reaction occurred as most abundant fraction (II), which was the major
well at 37°C as at 0°C when the antigen was removed sialoglycoprotein of red cell membranes, showed no
from the membrane. They suggested that at 37°C the or only weak I activity, but I active glycopeptides
antigen on the intact membrane may be “hidden,” could be isolated by digestion of fraction II with
whereas at 0°C the antigen may become available for trypsin. The major product of digestion, sialogly-
reaction with the antibody. Their studies led them to copeptide II T-2, showed I activity only after alkaline
believe that the difference between “I” and “i” is elimination of alkali-labile oligosaccharide chains.
largely a difference between the number of I antigenic Fraction III showed weak I activity but was slightly
sites. When the “affinity” of the antigen and antibody stronger than fraction II. Fraction III showed less MN
were increased by enzyme treatment of the RBCs or activity than fractions I and II.
by incubation in the cold, the total amount of antibody Lisowska and coworkers193 concluded that I active
fixed by both types of cells became nearly equal. receptors were present in all fractions of the RBC gly-
Gardas and Koscielak208 isolated an I active sub- coproteins they studied, but in some fractions they
stance from human RBCs by n-butanol extraction and were “masked” and could be exposed by enzymatic
found it to be identical to A, B, and H isolated anti- and chemical degradation of glycoproteins. The I
gens. The materials were sialic acid free and com- activity appeared to be associated with alkali-stable
prised about 90% carbohydrate, 7% to 7.5% of amino oligosaccharide chains. Sialic acid–rich alkali-labile
acids, and 2% of sphingosine. A, B, and H blood group oligosaccharides appeared to be responsible for the
activities were completely recovered in immune pre- steric hindrance for the reaction between anti-I and
cipitates of appropriate water-soluble antigens with some I antigens. The results were in favor of the
anti-I precipitating serum; on the contrary, I activity concept that a unique I substance is not present on
Specificity of Autoantibodies 249

RBCs but that I active receptors are located on differ- well developed at birth; the antibody was found to
ent ABH or MN-active molecules, in a position less or react better if the serum was at pH 6.5 or below (e.g.,
more available for reaction with anti-I. titer of 16 at pH 9.0 and 8000 at pH 6.2). In a series of
Several examples of ABO/I complex target antigens 268 cold autoantibodies investigated, two were
for nonpathogenic cold autoantibodies have been designated as anti-Sp1. Both of these were high titer
described (HI, Hi, IA, and IB). Two cases of CAS antibodies associated with AIHA; no examples of nat-
associated with autoanti-AI were described by urally occurring anti-Sp1 were found in healthy indi-
McGinniss.217,218 Postoway and coworkers219 viduals in the course of many thousand cold antibody
described a transient anti-IB in an A2B patient that investigations.
appeared to be the cause of the patient’s AIHA; this Independently, Roelcke222 described an antibody
antibody reacted to a titer of 2048 (at 4°C), and reacted called anti-HD (Heidelberg); this antibody appeared
up to 30°C with the patient’s RBCs. to be reacting with an antigen identical to Sp1. In 1969,
As the structure of ABH and Ii antigens became Roelcke published an extensive study showing het-
clear, it was no surprise that some antibodies erogeniety of the HD (Sp1) receptor.223 HD1 antigen
appeared to react with ABH–Ii complex antigens. could be demonstrated only on human RBCs, in con-
Wiener suggested that anti-I might be reacting against trast to the HD2 antigen, which could be demon-
the “nucleus” of ABO, and he has been proven to be strated on rat and guinea pig RBCs, as well as human
correct. Ii determinants are indeed internal structures RBCs. Neuraminidase and proteases inactivated both
of the ABH antigens. The Ii determinants are bound to antigens. However, quantitative differences were
glycolipids and glycoproteins in the RBC membrane; observed; the HD2 antigen showed more resistance to
carrier molecules are band 3, the major intrinsic mem- neuraminidase and proteases than HD1.223
brane protein and band 4.5 components. Only type 2 In 1970 Roelcke and Uhlenbruck224 suggested
chains [Galβ(1→4)GlcNAcβ(1→3)Gal-R] generate Ii replacing the terms “Sp” and “HD” with “Pr” (indi-
determinants on the RBC membranes. These type 2 cating the antigen inactivation by proteases). A
sequences are the basic structures of glycolipids of the complex heterogeneity has been discovered.221-229 Pr1
neolacto series. Ii antigens are built up by repeating antibodies were originally classified according to their
N-acetyllactosamine units: Anti-i recognizes the linear results with RBCs from humans and other animals.
chain and anti-I the branched chain. The branched Pr1h was said to be present only on human RBCs. Pr1d
chain is formed by adding a further lactosamine unit was said to be present on RBCs from human and some
in (1→6) linkage to the penultimate galactose (Gal) of animal species, including dogs. Pr2 had a similar
the linear chain. H substance is generated by substi- species distribution but in contrast to Pr1d, was found
tution of the terminal Gal of the I structure, with to be increased on dog RBCs and not destroyed by
Fucα(1→2). H substance serves as a precursor for A protease treatment of dog RBCs. The anti-SP1
and B. The fine specificity of anti-I and anti-i will described by Marsh and Jenkins is probably identical
depend on the extent of the Ii structure that reacts to anti-Pr1d or -Pr2 because SP1 was found to be inac-
with the specific antibody; many variations of the tivated by neuraminidase and to be present on guinea
theme can be encountered, thus explaining the many pig RBCs. The RBCs of 24,150 humans, tested by
subdivisions that have been made by serological Roelcke, were all found to have Pr1h and Pr2.
testing (e.g., ID, IF, IT, IH, IA, IB, iH, etc.). In 1971, a new determinant, Pra, was described.229
Two IgG cold autoagglutinins causing transient
“Cold” Autoantibody Specificities hemolytic anemia were detected in children; they
Other Than Anti-I and Anti-i reacted with all RBCs tested (cord and adult RBCs
reacted equally) and the reacting determinant was
Specificities other than anti-I or anti-i are rare.220 The inactivated by proteolytic enzyme, but in contrast to
most common specificity reacting with all human Pr1 and Pr2, it was not inactivated by neuraminidase. It
RBCs tested (adult and cord RBCs acting equally) was is easy to confuse anti-Pra with anti-EnaTS and anti-
first popularly called, rather facetiously, anti–“not-I.” EnaFS (see earlier). Another determinant of the Pr
Dr. Marsh, who christened it so, told us he was cha- system was also described in 1971, Pr3.228 A mono-
grined that the name ever appeared in print, and clonal IgM (kappa) antibody, associated with CAS,
called it anti-SP1 when the first extensive serological occurring after rubella infection, was shown to react as
data on this antibody was published.221 an anti-Pr, but was different than anti-Pr1, -Pr2, and
Marsh and Jenkins221 called the antibody anti-Sp1 -Pra. Pr3 determinants are found on cat and sheep RBCs
(species) as no human RBCs lacking the reacting which lack Pr1 and Pr2 determinants. By carbodiimide
antigen could be found but no RBCs from 25 other treatment of human red cell glycoproteins, which
species reacted. The Sp1 antigen showed a striking causes intramolecular coupling of N-acetylneuraminic
difference from I and i in that the Sp1 antigen could be acid carboxyl group and nucleophilic centers of the
destroyed or markedly reduced by treating the RBC glycoprotein backbone, Pr3 antigen activity was greatly
with proteolytic enzymes (trypsin, bromelin, papain, increased, whereas Pr1 and Pr2 were inactivated.
and ficin); both I and i give enhanced reactions follow- The Pr determinants are present on RBC gly-
ing enzyme treatment. The antigen was found to be cophorins (e.g., glycophorin A). The O-glycosidically
250 Immune Hemolytic Anemias

linked oligosaccharides of glycophorins are the Pr that clearly differed and because these specificities
determinants. All fragments of glycophorin A carry- correspond to chemical differences in the RBC mem-
ing oligosaccharides are Pr1-, Pr2-, and Pr3-inactive. brane, Wang and coworkers241 suggested that their
The fine structure of the Pr antigenic site is unknown study illustrated a direct correlation between anti-
at present. Because En(a–) RBCs lack the major gly- body specificity and the structure of the light and
cophorin, glycophorin A, they react very poorly, or heavy chain variable regions. The case “Rob” that we
not at all, with anti-Pr. As mentioned earlier, En(a–) described237 had an interesting history. The patient
RBCs adsorb anti-Pr; thus, glycophorins other than was a 51-year-old white man whose major symptoms
glycophorin A may carry some Pr. MkMk RBCs that consisted of extreme sensitivity to the cold, as mani-
lack glycophorins A and B do not react with anti-Pr, fested by bluish discoloration of the extremities, ears,
but adsorption and elution studies have not been and face. With intense exposure to cold, his entire
performed. appearance was purplish, and his hands and, in par-
Pr antibodies can now be classified biochemically.220 ticular, his feet became extremely painful. The diagno-
Anti-Pr1h and anti-Pr3h react exclusively with human sis of cold agglutinin syndrome was not made until 21
RBCs. Anti-Pr1d and anti-Pr3d react with human and years later. At this time, electrophoresis of his serum
dog RBCs. Pr2 is not limited to human RBCs. showed a monoclonal spike in the β globulin region,
Antibodies (anti-PrM and -PrN) acting like anti-Pr but which was shown by immunoelectrophoresis to be
showing a preference for RBCs of M or N phenotype, IgA. The bone marrow was hypercellular and con-
respectively, have been described.220 Pr1, Pr2, and Pr3 tained approximately 10% mature plasma cells. A
can be differentiated by simple biochemical proce- diagnosis of multiple myeloma was considered,
dures, using periodate-oxidized and carbodiimide- although the patient did not develop osteolytic bone
treated glycophorins. Pr1 is inactivated by both lesions or urine protein abnormalities. Physical exam-
procedures; Pr2 is increased 100- to 200-fold by oxida- ination revealed no lymphadenopathy, splenomegaly,
tion and is inactivated by carbodiimide treatment; or hepatomegaly. Bone marrow examinations over the
Pr3 is increased 100- to 200-fold by carbodiimide- next 5 years showed an increase in plasma cells, and
treatment and is inactivated by oxidation. When char- the serum IgA level rose to a concentration of 1.01 g%.
acterizing 32 anti-Pr cold autoagglutinins, Roelcke220 His cold agglutinin titre was 16,000, reacting up to
found 24 anti-Pr1h, 3 anti-Pr2, and 5 anti-Pr3. 35°C. The serum caused no hemolysis in vitro of
Several reports of anti-Pr cold autoantibodies associ- normal or enzyme-treated RBCs. The antibody being
ated with AIHA have appeared in the literature.228-234 IgA was incapable of activating complement; thus,
Three were unusual cases associated with anti-Pra. although the patient had clinical signs due to the pow-
Curtis and coworkers232 reported a life-threatening, erful cold autoagglutinin (e.g., acrocyanosis), no evi-
DAT-negative acute hemolytic anemia associated with dence of hemolytic anemia was ever detected.
a non–complement-activating IgG1 monoclonal Target Antigens (Gd, Fl, Vo, Li) That Are Sialidase
(kappa) cold autoantibody with anti-Pra specificity. Sensitive and Protease Resistant. Gd antigens are
Kadota and coworkers233 reported a hemolytic anemia glycolipid dependent (Gd) and are fully expressed on
associated with a rubella infection and anti-Pra. Ramos adult and cord RBCs. They are created by sialyation of
and coworkers234 reported a refractory immune Ii glycolipid antigens.220 The structure of the Gd
hemolytic anemia asssciated with a high thermal determinant is NeuNAc(2→3)[Gal(1→4)GlcNAc(1
amplitude, low affinity IgG anti-Pra cold autoantibody. → 3)n]. Glycophorins are Gd inactive. The Gd major
It is interesting that only five examples of IgA high active ganglioside, sialylneolactotetraosylceramide, is
titer cold autoagglutinins have been reported,235-239 increased in the RBC membranes of p individuals. It
and four have been shown to be monoclonal (kappa) has been suggested that antibodies designated anti-p
and have anti-Pr1 specificity. Most anti-I associated may be anti-Gd.220
with chronic CAS are IgM monoclonal (kappa) pro- Anti-Fl may be confused with anti-I as the Fl
teins, but rare examples of IgM (lambda) cold agglu- antigen is fully expressed only on Iadult RBCs; the anti-
tinins have been reported.225 Three of these were gen is poorly expressed on icord and iadult RBCs. Anti-
originally reported as having anti–“not-I” specificity, Fl differs from anti-I only in not reacting with
and one of them has since been identified as sialidase-treated RBCs. The branched I active neolac-
anti-Pr1.224 The restricted nature of anti-I cold agglu- tosequence is the basic structure of the Fl antigen, but
tinins is not limited to their constant regions; their one or both branches are sialylated and NeuNAc is
light chain variable regions are predominantly VKII the immunodominant component.220 I active sialogly-
subgroup, and their heavy chain variable regions are coproteins will inhibit antiFl.220 Fl-like I antigens are
predominantly VHI subgroup.240,241 Wang and membrane glycolipids and glycoproteins. Oh RBCs
coworkers241 studied the amino acid sequence of the have very reduced reactivity with anti-Fl.220
IgA cold agglutinin “Rob” that we had reported as Vo and Li are targets for cold autoagglutinins that
having anti-Pr specificity in 1973.237 They were able to are protease-resistant and fully expressed only on icord
define a new kappa chain variable region subgroup and iadult RBCs; thus, anti-Vo and -Li may be confused
that was designated VKIV. Because the anti-Pr and with anti-i. Unlike i, Vo and Li are sialidase sensitive.
anti-I cold agglutinins had variable region subgroups Vo and Li differ in their susceptibility to sialidase
Specificity of Autoantibodies 251

treatment. Li is susceptible to sialidase on an intact similarly, M, N, Ena, Wrb, and Pr are on the same
surface, whereas Vo is susceptible to sialidase only structure (i.e., glycophorin A), it should be no surprise
after protease treatment. It has been suggested that Vo that interpretation of specificity, based on serological
and Li are sialylated linear type 2 (neolacto) chains, reactions, can be confusing and inaccurate.
NeuNAc(2→3)[Gal(1→4)Glc-Nac(1→3)], that is, sialy- Tables 7-13 and 7-14 show the serological character-
lated i determinants.220 istics of the antibodies discussed above.
Target Antigens (Lud, Sa) That Are Sialidase- Roelcke220 has suggested a new terminology for Fl,
Sensitive and Partially Inactivated by Proteases. Vo, and Gd. The antigens share several characteristics:
Another cold autoagglutinin, anti-Lud, may also be
confused with anti-I and anti-Fl. Lud is fully 1. The antigens are differentiation antigens. Sia-b1
expressed on Iadult but not on cord RBCs; thus, if only (formerly F1) is fully expressed only on adult RBCs.
adult and cord RBCs are used, anti-I, -Fl, and -Lud Sia-l1 (formerly Vo) is fully expressed only on
give similar results. Unlike I and Fl, Lud is fully newborn RBCs. As an exception, Sia-lb1 (formerly
expressed on iadult RBCs. Proteases partially inactivate Gd) is expressed on adult and newborn RBCs in
Lud but do not inactivate I or Fl.220 Thus, anti-Lud equal strength.
recognizes sialidase-sensitive antigens fully expressed 2. The antigens are present on the rare adult RBCs
only on adult RBCs. The structure of Lud is unknown with the i phenotype (i adult RBCs) in equal
at present. strength as on newborn (i cord) RBCs. It can, there-
The Sa antigens are sialidase sensitive but only par- fore, be concluded that the structures responsible
tially inactivated by papain and do not react with gly- for Ii and Sia-1, -b, -1b antigens are related.
coproteins obtained from papain-treated RBCs. The 3. The antigens are inactivated by sialidase treatment
antigens are fully expressed on adult and cord RBCs. of RBCs, indicating that NeuNAc, not involved in Ii
Glycophorin A carries Sa determinants. Like Pr2 (see antigenic determinants, serves as immunodomi-
later), Sa antigens are gangliosides, but unlike Pr2 they nant component.
are restricted to the neolacto series.220 Anti-Sia-b1 (anti-Fl) may initially be mistaken for
It is obvious from the above data that most reports anti-I. Anti-Sia-l1 (anti-Vo) and anti-Sia-lb1 (anti-
of specificity of cold autoagglutinins being anti-I or Gd) may initially be mistaken for anti-i (Table 7-15).
anti-i are now suspect as these sera have not usually
been tested with sialidase-treated RBCs. Indeed, this
is a technical problem as most sera contain anti-T that
MISCELLANEOUS TARGETS
may react with sialidase-treated RBCs. It is possible FOR COLD AUTOANTIBODIES
that sera designated as anti-I or anti-i may be found to
contain anti-Gd, -Fl, -Vo, -Li, or -Lud, together with Rare examples of anti-A and anti-B have been
anti-I/i or even alone. This would parallel the discov- reported as cold and warm autoantibodies.242 Three
ery that anti-“dl” could contain, or be, anti-Ena or examples of autoanti-A and one example each of
-Wrb. As the epitopes for A, B, H, I, i, Gd, Fl, Vo, Li, autoanti-IB and autoanti-B have been reported to be
and Lud are all on the same basic structure, and, associated with CAS.242-244 Sokol and coworkers242

TABLE 7-13. SEROLOGIC CHARACTERISTICS OF


AUTOANTIBODIES THAT REACT WITH TARGET ANTIGENS
THAT MAY BE AFFECTED BY PROTEASES OR SIALIDASES

Iadult Iadult Iadult Iadult


Antibody Iadult icord iadult PT ST En(a-) Wr(b-)

Anti-I + ↓ ↓ ↑ ↑ + +
-i ↓ + + ↑* ↑* ↓ ↓
-IT ↓ + ↓ + + ↓ ↓
-Pr + + + 0 0 0** 0
-Pra + + + 0 + 0** 0
-Gd + + + + 0 + +
-Fl + ↓ ↓ + 0 + +
-Vo ↓ + + ↑* ↓* + +
-Li ↓ + + +* 0* + +
-Lud + ↓ + ↓ 0 + +
-Sa + + + ↓ 0 + +
-Ena + + + 0 + 0 0

PT, protease (papin or ficin) treated; +, antibody reacts; ↑, increased reaction compared with
same RBCs untreated; ST, sialidase (neuraminidase) treated; 0, antibody does not react;
↓, decreased reaction; *, reactions apply to icord and iadult, but not Iadult RBCs;
**, antibody may be adsorbed/eluted by or from RBCs.
252 Immune Hemolytic Anemias

TABLE 7-14. TYPICAL REACTIONS OF ANTI-Pr SUBSPECIFICITIES§

Antibody Directed Against

Red Blood Cells Pr1h Pr1d Pr2 Pr3h Pr3d Pra PrM PrN

Iadult
Untreated + + + + + + +‡ +⎪⎪
Protease 0 0 0 0 0 0 0 0
Sialidase 0 0 0 0 0 + 0 0
iadult
Untreated + + + + + + +‡ +⎪⎪
Protease 0 0 0 0 0 0 0 0
Sialidase 0 0 0 0 0 + 0 0
icord
Untreated + + + + + + +‡ +⎪⎪
Protease 0 0 0 0 0 0 0 0
Sialidase 0 0 0 0 0 + 0 0
Dog
Untreated 0 +† + 0 + 0
Protease 0 V E 0 0 0
Sialidase 0 0 0 0 0 0
Sheep 0 0 0 + +
Cat 0 0 0 + +
Guinea Pig V V + E
Rat V V V V
Rabbit¶ 0 0 0 0

+, Agglutination: V, variable reaction (i.e., some negative, some positive); in titration studies, results with red
blood cells (RBCs) marked V usually react to a lower titer than those marked +; E, enhanced reaction; in titration
studies, results with RBCs marked E usually react to higher titers than those marked +; blank spaces, no
information available.
† Some anti-Pr
1d react only with dog RBCs.
‡ Reactions of all RBCs equal in tests at low (4°C) temperatures. Reactions of M+ RBCs stronger than those of
M– RBCs in tests at higher (20°–25°C) temperatures.
§ The most usual reactions are shown.
⎪⎪ Similar to above except that preference is for N+ RBCs.
¶ RBCs from rabbits are useful in differential adsorption studies because they lack the Pr determinants but carry I
and i.

reported six cases of autoanti-A detected in a 32-year total bilirubin = 1.9 mg/dL). An IgM kappa mono-
period (4668 patients with autoantibodies studied). clonal protein was present. Regular plasmapheresis
Three of the autoanti-A caused hemolytic anemia treatments were helpful in relieving clinical symp-
associated presumably with CAS. The autoanti-B was toms. The patient’s RBCs were sensitized with IgM
detected in a 52-year-old group B man who presented (3+), IgA (1+), and C3 (3+). The clinical laboratory
with numbness, tingling, and blue discoloration of reported a normal cold agglutinin titer of 2 (i.e., group
fingers and toes when exposed to cold (acro- O RBCs were used), but later the serum was shown to
cyanosis).244 Later, the patient had slurring of speech contain autoanti-B reacting to a titer of 8192 at 4°C and
and headaches; magnetic resonance angioplasty/ 16 at 30°C (nonreactive at 37°C) with allogeneic RBCs.
magnetic resonance imaging showed mild small DTT-treated autologous RBCs reacted to a titer of 8192
vessel brain disease. Hemolytic anemia was not at 4°C and reacted at 30°C, in the presence of albumin.
apparent (hemoglobin/hematocrit = 13.1 g/dL/39.6%, The antibody caused weak hemolysis of group B

TABLE 7-15. EXPRESSION OF SIA-B1, SIA-l1, SIA-lB1 ANTIGENS COMPARED WITH Ii ANTIGENS
ON HUMAN UNTREATED AND ENZYME-TREATED RBCs

Antigen Designation untr. pap. sial. endo. untr. pap. sial. pap/sial endo. untr.

I + ↑ ↑ ↓ ↓ ↓ ↓
Sia-b1 F1 + + – + ↓ ↓ ↓
i ↓ + ↑ ↑ ↑ – + +*
Sia-Il Vo ↓ + ↑ + – – + +*
Sia-Ib1 Gd + + – + + + – – + + +

untr., untreated; pap., papain treated; sial., sialidase treated; endo., endo-β-galactosidase treated; pap./sal., sialidase treated after papainization;
+, optimal reaction with untreated RBCs; ↑, reaction (slightly) increased; ↓, reaction markedly decreased; – no reaction; *, expressed as on adult
RBCs.
Specificity of Autoantibodies 253

enzyme-treated but not untreated RBCs. A heat eluate 22°C to 27°C; no reactions occurred at 37°C. One very
from the patient’s RBCs reacted with group B enzyme- unusual patient with an IgM autoanti-D that reacted
treated but not untreated RBCs. As the autoanti-B optimally in the cold, but reacted up to 37°C, was
reacted up to 30°C in vitro, activated complement [in described by Longster and Johnson.255
vitro (enzyme-treated B RBCs were hemolyzed) and
in vivo (i.e., patient’s RBCs were strongly sensitized OPTIMAL REACTIONS WITH STORED
with C3)], and the clinical symptoms indicated that
the antibody was reacting in vivo, it was hard to OR “OLD” RBCs
understand why this patient did not have a more
obvious hemolytic anemia.244 In 1952 Brendemoen and coworkers153 described a
Anti-M and -N have been reported as pathogenic cold autoagglutinin that reacted with stored RBCs at
and nonpathogenic cold autoantibodies. Two cases of 5°C and 15°C but not 37°C, and not with fresh RBCs at
AIHA associated with high titer, high thermal ampli- any temperature. No clinical data on the patient were
tude autoanti-M245,246 and one case of autoanti-N247 given. Stratton154 described a similar antibody, but
have been reported. Many cases of nonpathogenic this antibody reacted better at 37°C. In 1961 Jenkins
autoanti-M and -N have been detected27; these and Marsh155 described three patients with AIHA
include autoanti-N in dialysis patients that may be associated with autoagglutinins reacting with stored
induced by formalin.27 RBCs but not fresh RBCs. The serology of only one
In 1980 the sera of two patients with CAS were patient was reported in detail. The DAT was positive,
found to contain antibodies of a new specificity.248 due to complement sensitization. The autoagglutinin
These IgM autoagglutinins reacted equally well with reacted equally well at 4°C, 18°C, and 37°C. The anti-
Iadult and iadult RBCs and weaker with cord RBCs; they body would react with RBCs that had been enzyme-
reacted well with sialidase- and protease-treated or heat (56°C)-treated. After the patients were treated
RBCs. They appeared to react better at room tempera- with steroids the antibodies disappeared in two cases,
ture than at 37°C or 4°C. As they were inhibited by and in one case the remaining antibody reacted only at
urine from Sd(a+) but not Sd(a–) individuals and did low temperatures; the latter finding leads one to
not appear to be anti-Sda, they were called anti-Sdx.248 suspect this was a high thermal amplitude “cold” anti-
They were later renamed anti-Rx; the inhibition with body rather than a “warm” autoantibody.
Sd(a+) urine was found to be a serologic artifact.249 Ozer and Chaplin156 studied an IgM monoclonal
Severe AIHA, including a fatality, associated with antibody, with similar specificity, in great detail. Their
autoanti-Rx, has been reported.248,250,251 It is interest- patient had macroglobulinemia and hemolytic
ing to note that 12 cases were reported during the 1980 anemia. The antibody had a cold agglutinin titer of
influenza epidemic in New York.248-250 100,000 to 500,000 and reacted up to 37°C. Like
Two “new” target antigens for cold autoantibodies Stratton and coworkers,154 Ozer and Chaplin156 noted
were named Me and Ju by Salama and coworkers252 that microscopically the agglutination gave a mixed-
and Göttsche and coworkers.253 Anti-Me was detected field appearance. Red cell survival studies were
in a patient with AIHA and Waldenström’s macroglo- carried out using fresh and stored RBCs. Fresh RBCs
bulinemia. The antibody was IgM (kappa), of moderate survived normally and stored RBCs (mean storage
titer (128 at 0°C), reacting up to 30°C with adult and period of 30.5 days) survived poorly (71% at 1 hour,
cord RBCs. The antibody reacted better with protease- 53% at 6 hours, 48% at 24 hours, 39% at 48 hours).
and sialidase-treated RBCs; papain-treated adult and Beaumont and coworkers256 described an antibody
cord RBCs were hemolyzed at 20°C and 37°C. The anti- in a patient with macroglobulinemia, but no hemolytic
body was named anti-Me (milk-enhanced) because it anemia. This IgM antibody reacted with stored RBCs
gave greatly enhanced reactions in the presence of pre- but not fresh RBCs, and also precipitated known low-
heated human milk (titer of 1024 with milk versus 16 density lipoprotein (LDL). It is of interest to note that
without milk at 20°C).252 A cold autoantibody causing LDL has been identified as an autoantigen in the AIHA
CAS was shown to have characteristics that seemed to of NZB mice.257 Lightwood and Scott258 reported a case
exclude anti-I, -i, -Pr, -Gd, -Sa, -Vo, -Li, -Fl, -Lud, and of CAS, with high titer anti-I and anti-i, where an IgM
-Me.253 The antibody reacted equally well with adult antibody to stored RBCs developed 5 years after the
and cord RBCs. The antigen reactivity was strongly initial diagnosis of AIHA. A cold autoantibody against
diminished by sialidase (neuraminidase) treatment and stored RBCs was reported by Easton and coworkers157
to a lesser degree by protease (papain) treatment. The in a patient with mild hemolytic anemia, cirrhosis of
target antigen was named Ju.253 the liver, and a false-positive test for syphilis.
An interesting finding was that of Perrault, who As discussed for WAIHA, reticulocytopenia has also
found 10 examples of cold-reacting autoanti-LW in been observed in AIHA associated with “cold” autoan-
45,000 donors, whose sera were screened using a low tibodies.259 In 1990 Kesmin and colleagues260 presented
ionic strength Polybrene AutoAnalyzer system.254 a case of CAS where the high titer, high thermal ampli-
One of the antibodies was IgM, the others were tude cold agglutinin appeared to react preferentially
thought to be IgG. The antibodies reacted well up to with younger (i.e., reticulocyte-rich) RBCs. This was
20°C; a marked decrease in reactivity was noted at noticed because clumps on the peripheral blood smear
254 Immune Hemolytic Anemias

appeared to be mainly polychromatic RBCs. Cold 7. Dacie JV, Cutbush M: Specificity of auto-antibodies in acquired
agglutinin titers/scores at 20°C with reticulocyte-rich haemolytic anaemia. J Clin Pathol 1954;7:18.
8. Wiener A, Gordon EG, Gallop C: Studies on autoantibodies in
RBCs were 32/54 and 8/34 with reticulocyte-reduced human sera. J Immunol 1953;71:58.
RBCs. Thus, the reactions did not reflect the presence or 9. Pirofsky B, Pratt K: The antigen in autoimmune hemolytic
absence of a specific target antigen, but perhaps the anemia. I. Reactivity of human autoantibodies and rhesus anti-
presence of more antigen on younger RBCs. bodies with primate and non-primate erythrocytes. Am J Clin
Pathol 1966;45:75-81.
10. Dacie J: The Haemolytic Anaemias, 3rd ed. vol. 3, The
AUTOANTIBODY SPECIFICITY Auto-Immune Haemolytic Anaemias. New York: Churchill
Livingstone, 1992:161.
ASSOCIATED WITH PAROXYSMAL 11. van Loghem JJ, van der Hart M: Varieties of specific auto-anti-
COLD HEMOGLOBINURIA bodies in acquired haemolytic anaemia. Vox Sang 1954;4:2.
12. Flückiger P, Ricci C, Usteri C: Zur Frage der Blutgruppens-
pezifität von Autoantikörpern. Acta Haematol 1955;13:53.
In 1963 Levine and coworkers261 reported that three 13. Dausett J, Colombani J, Jean RG, et al: The serology and
prognosis of 128 cases of autoimmune hemolytic anemia. Blood
sera from six patients with paroxysmal cold hemoglo- 1954;14:1280.
binuria (PCH) failed to react with the rare Tj(a–) RBCs, 14. Weiner W, Vos GH: Serology of acquired hemolytic anemias.
but reacted with all other RBCs tested. Thus, Levine Blood 1963;22:606–613.
and coworkers261 suggested the specificity of the 15. Vos GH, Petz LD, Fudenberg HH: Specificity of acquired
autoantibodies in PCH was anti-P+P1 (anti-Tja). haemolytic anaemia autoantibodies and their serological char-
acteristics. Br J Haematol 1970;19:57–66.
Worlledge and Rousso262 extended these findings by 16. Vos GH, Petz LD, Fudenberg HH: Specificity and
testing 11 patients with PCH, using the rare Pk RBCs in immunoglobulin characteristics of autoantibodies in acquired
addition to p [Tj(a–)] RBCs (Levine and coworkers had hemolytic anemia. J Immunol 1971;106:1172–1176.
mentioned in an addendum to their article that three of 17. Vos GH, Petz LD, Garratty G, Fudenberg HH: Autoantibodies
in acquired hemolytic anemia with special reference to the LW
their sera tested did not react with Pk RBCs). All 11 sera system. Blood 1973;42:445–453.
reacted with P1 and P2 RBCs but not with p or Pk RBCs, 18. Dacie JV, Worlledge SM: Auto-immune hemolytic anemias.
which suggested a specificity similar to that of the Prog Hematol 1969;6:82–119.
anti-P naturally occurring in the rare Pk individuals. 19. Leddy JP, Peterson P, Yeaw MA, Bakemeier RF: Patterns of
Other workers have also since reported cases of PCH serologic specificity of human γG erythrocyte autoantibodies. J
Immunol 1970;105:677–686.
with P specificity.263-267 20. Rosse WF: Fixation of the first component (C’1a) by human
Both von dem Borne and coworkers268 and Ramos antibodies. J Clin Invest 1968;47:2430.
and coworkers269 described cases of non-Hodgkin’s 21. Sokol RJ, Hewitt S, Booker DJ, Bailey A: Red cell autoantibod-
lymphoma associated with an autoimmune hemolytic ies, multiple immunoglobulin classes, and autoimmune
hemolysis. Transfusion 1990;30:714–717.
anemia that did not appear to be CAS but were not 22. Dacie JV: Autoimmune hemolytic anemia. Arch Int Med
typical PCH; one autoantibody was a monoclonal IgM 1975;135:1293.
(kappa) anti-P, and the other was a mixture of anti-P 23. Issitt PD, Pavone BG, Goldfinger D, et al: Anti-Wrb, and other
and anti-ITP cold hemolysins. autoantibodies responsible for positive direct antiglobulin
There are a few reports of other specificities said to tests in 150 individuals. Br J Haematol 1976;34:5–18.
24. Celano MJ, Levine P: Anti-LW specificity in autoimmune
be associated with PCH; these include single case acquired hemolytic anemia. Transfusion 1967;7:265–268.
reports of anti-p,270 anti-HI,271 anti-I,272 anti–“Pr- 25. Marsh WL, Reid ME, Scott EP: Autoantibodies of U blood
like,”273 and several anti-i.274-276 group specificity in autoimmune haemolytic anaemia. Br J
All cases of PCH we have encountered in the past Haematol 1989;72:625.
26. Sistonen P, Tippett P: A “new” allele giving further insight into
30 years were associated with anti-P specificity. Sokol the LW blood group system. Vox Sang 1982;42:252–255.
and coworkers277 found anti-P specificity in 27 of 30 27. Issitt PD, Anstee DJ: Applied blood group serology, 4th ed.
patients with PCH; specificity was not clearly defined Durham, NC: Montgomery Scientific Publications, 1998.
in the other three cases. 28. Eyster ME, Jenkins DE, Jr: IgG erythrocyte autoantibodies:
Comparison of in vivo complement coating and in vitro “Rh”
specificity. J Immunol 1970;105:221–226.
29. Nugent ME, Colledge KI, Marsh WL: Autoimmune hemolytic
R E F E R E N C E S anemia caused by anti-U. Vox Sang 1971;20:519–525.
30. Schmidt PJ, Lostumbo MM, English CT, Hunter OB, Jr: Aberrant
1. Denys P, van den Broucke J: Anémie hémolytique acquise et U blood group accompanying Rhnull. Transfusion 1967;7:33–34.
réaction de Coombs. Arch Franc Pédiatr 1947;4:205. 31. Worlledge SM: A classification of AIHA, based on clinical
2. Kuhns WJ, Wagley PF: Hemolytic anemia associated with grounds and the use of antiglobulin sera specific for
atypical hemagglutinins. Ann Intern Med 1949;30:408. immunoglobulin class and various complement components,
3. Race RR, Sanger R, Selwyn JG: Possible deletion in human Rh and its use in clinical practice. In Proceedings of the 25th
chromosome: A serological and genetical study. Br J Exp Annual Meeting of the American Association of Blood Banks
Pathol 1951;32:124. and the 13th Congress of the International Society for Blood
4. Weiner W, Battey DA, Cleghorn TE, et al: Serological findings Transfusion, 1972:43 (Abstr).
in a case of haemolytic anaemia. Br Med J 1953;2:125. 32. Goldfinger D, Zwicker H, Belkin GA, et al: An autoantibody
5. Holländer L: Specificity of antibodies in acquired haemolytic with anti-Wrb specificity in a patient with warm autoimmune
anaemia. Experientia 1953;9:468. hemolytic anemia. Transfusion 1975;15:351–352.
6. Dacie JV: Serology of acquired haemolytic anaemia. 33. Bell CA, Zwicker H, Sacks HJ: Autoimmune hemolytic
Proceedings of 4th Congr Europ Soc Haemat, Amsterdam, anemia: Routine serologic evaluation in a general hospital
1953. population. Am J Clin Pathol 1973;60:903–911.
Specificity of Autoantibodies 255

34. Garratty G, Brunt D, Greenfield B, et al: An autoanti-Ena mim- 58. Garratty G, Sattler MS, Petz LD, Flannery EP: Immune
icking an alloanti-Ena associated with pure red cell aplasia. hemolytic anemia associated with anti-Kell and a carrier state
Transfusion 1983;33:408 (Abstr). for chronic granulomatous disease. Blood Transfus Immuno-
35. D’Orsogna DE, Heinz R, Wawryszczuk V, Loftis LL, Harrison haematol 1979;22:529.
CR: Severe autoimmune hemolytic anemia with anti-Ena 59. Giblett ER, Klebanoff SJ, Pincus SH, et al: Kell phenotypes in
specificity in a four week old infant girl. Transfusion chronic granulomatous disease: A potential transfusion
1991;31:23S (Abstr). hazard. Lancet 1971;i:1235–1236.
36. Garratty G, Arndt P, Domen R, et al: Severe autoimmune 60. Marsh WL, Øyen R, Nichols ME, et al: Chronic granulomatous
hemolytic anemia associated with IgM warm autoantibodies disease and the Kell blood groups. Br J Haematol 1975;
directed against determinants on or associated with gly- 29:247–262.
cophorin A. Vox Sang 1997;72:124–130. 61. Wilkinson SL, Vaithianathan T, Issitt PD: On the high inci-
37. Dankbar DT, Pierce SR, Issitt PD, et al: Fatal intravascular dence of anti-HL-A antibodies in anti-D typing reagents.
hemolysis associated with autoanti-Wrb. Transfusion Illustrated by a case of Matuhasi-Ogata phenomenon mimick-
1987;27:534 (Abstr). ing a “D with anti-D” situation. Transfusion 1974;14:27–33.
38. Ainsworth BM, Fraser ID, Poole GD: Severe haemolytic 62. Issitt PD, Zellner DC, Rolih SD, Duckett JB: Autoantibodies
anaemia due to anti-Wrb. In Book of Abstracts from the Joint mimicking alloantibodies. Transfusion 1977;17:531–538.
Meeting of 20th Congress of International Society of Blood 63. Viggiano E, Clary NL, Ballas SK: Autoanti-K antibody mim-
Transfusion/British Blood Transfusion Society, 1988:82. icking an alloantibody. Transfusion 1982;22:329–332.
39. Issitt PD, Combs MR, Allen J, et al: Anti-Dib as a red cell 64. Puig N, Carbonell F, Marty ML: Another example of mimick-
autoantibody. Transfusion 1996;36:802–804. ing anti-Kpb in a Kp(a+b-) patient. Vox Sang 1986;51:57–62.
40. Sherburne B, Salamon JL, Shirey RS, et al: Warm autoimmune 65. Manny N, Levene C, Sela R, et al: Autoimmunity and the Kell
hemolytic anemia (AIHA) associated with IgG anti-Pr1h. blood groups: Autoanti-Kpb in a Kp(a+b–) patient. Vox Sang
Transfusion 1987;27:535 (Abstr). 1983;45:252–256.
41. Dube VE, House RF, Jr, Moulds J, Polesky HB: Hemolytic anemia 66. Win N, Kaye T, Mir N, Damain-Willems C, Chatfield C:
caused by autoanti-N. Am J Clin Pathol 1975;63:828–831. Autoimmune haemolytic anaemia in infancy with anti-Kpb
42. Cohen DW, Garratty G, Morel P, Petz LD: Autoimmune specificity. Vox Sang 1996;71:187–188.
hemolytic anemia associated with IgG autoanti-N. Transfusion 67. Marsh WL, DiNapoli J, Øyen R: Auto-immune hemolytic
1979;19:329–331. anemia caused by anti-K13. Vox Sang 1979;35:174–178.
43. Combs MR, Telen MJ, Hall SE, Rosse WF: A case report: IgG 68. Marsh WL, Oyen R, Alicea E, et al: Autoimmune hemolytic
autoanti-N as a cause of severe autoimmune hemolytic anemia and the Kell blood groups. Am J Hematol
anemia. Immunohematology 1990;6:83. 1979;7:155–162.
44. Garratty G, Arndt P, Tsuneta R, Kanter M: Fatal hemolytic 69. Sabo B, Keeling MM, Winkler MA, McCreary J: Coexistence of
anemia associated with autoanti-N. Transfusion 1994;34:20S a high frequency Kell-related autoantibody and alloanti-Kpb in
(Abstr). a case of autoimmune disease (Abstr). In Book of Abstracts
45. Crosson JT, Moulds J, Comty CM, Polesky HF: A clinical study from the Joint Meeting of the 19th Congress of the
of anti-NDP in the sera of patients in a large repetitive International Society of Haematology/17th Congress of the
hemodialysis program. Kidney Int 1976;10:463–470. International Society of Blood Transfusion, 1982:235.
46. Fassbinder W, Frei U, Koch KM: Haemolysis due to formalde- 70. Vengelen-Tyler V, Gonzalez B, Garratty G, et al: Acquired loss
hyde-induced anti-N-like antibodies in haemodialysis of red cell Kell antigens. Br J Haematol 1987;65:231–234.
patients. Klin Wochenschr 1979;57:673–679. 71. Eveland D: AutoantiJsb enhanced by polyethylene glycol
47. Johnson MH, Plett MJ, Conant CN, Worthington M: (Abstr). In Book of Abstracts from the ISBT/AABB Joint
Autoimmune hemolytic anemia with anti-S specificity. Congress. Arlington, VA: American Association of Blood
Transfusion 1978;18:389 (Abstr). Banks, 1990:156.
48. Alessandrino EP, Costamagna L, Pagani A, Coronelli M: Late 72. Holmes LD, Pierce SR, Beck M: Autoanti-Jka in a healthy blood
appearance of autoantibody anti-S in autoimmune hemolytic donor. Transfusion 1976;16:521.
anemia. Transfusion 1984;24:369–370. 73. Patten E, Beck CE, Scholl C, et al: Autoimmune hemolytic
49. Domen RE, Clarke A: Case reports: Red blood cell autoanti- anemia with anti-Jka specificity in a patient taking Aldomet.
bodies mimicking alloantibodies. Immunohematology Transfusion 1977;17:517–520.
1991;7:98. 74. Guillausseau PJ, Wautier JL, Boizard B, et al: Une variété rare
50. Reynolds MV, Vengelen-Tyler V, Morel PA: Autoimmune d’anémie hémolytique auto-immune: la spécificité anti-Jka.
hemolytic anemia associated with autoanti-Ge. Vox Sang Semin Hop Paris 1982;58:803.
1981;41:61–67. 75. Hoffman M, Berger MB, Menitove JE: Autoimmune hemolytic
51. Beattie KM, Sigmund KE: A Ge-like autoantibody in the serum anemia due to autoanti-Jka. Lab Med 1982;13:674.
of a patient receiving gold therapy for rheumatoid arthritis. 76. Strikas R, Seifert MR, Lentino JR: Autoimmune hemolytic
Transfusion 1987;27:54–57. anemia and Legionella pneumophilia pneumonia. Ann Intern
52. Gottsche B, SalamaA, Mueller-Eckhardt C: Autoimmune- Med 1983;99:345.
hemolytic anemia associated with an IgA autoanti-Gerbich. 77. Ciaffoni S, Ferro 1, Potenza R, Campo G: Evans’s syndrome: A
Vox Sang 1990;58:211–214. case of autoimmune thrombocytopenia and autoimmune
53. Poole J, Reid ME, Banks J, et al: Serological and immunochem- hemolytic anemia caused by anti-Jka. Haematologica
ical specificity of a human autoanti-Gerbich-like antibody. Vox 1987;72:245–247.
Sang 1990;58:287–291. 78. Sander RP, Hardy NM, van Meter SA: Anti-Jka autoimmune
54. Shulman IA, Vengelen-Tyler V, Thompson JC, et al: Autoanti- hemolytic anemia in an infant. Transfusion 1987;27:58–60.
Ge associated with severe autoimmune hemolytic anemia. Vox 79. Ganly PS, Laffan MA, Owen 1, Hows JM: Auto-anti-Jka in
Sang 1990;59:232–234. Evans’s syndrome with negative direct antiglobulin test. Br J
55. Sererat T, Veidt D, Arndt PA, Garratty G: Warm autoimmune Haematol 1988;69:537–539.
hemolytic anemia associated with autoanti-Ge. 80. Alpaugh K, Sysko-Stein L, Lusch C, et al: Autoanti-Jka associ-
Immunohematology 1998;11:26. ated with hemolysis after transfusion with Jk(a+) RBCs. Lab
56. Seyfried H, Gorska B, Maj S, et al: Apparent depression of anti- Med 1989;20:682–684.
gens of the Kell blood group system associated with autoim- 81. McGinniss MH, Leiberman R, Holland PV: The Jkb red cell
mune acquired hemolytic anaemia. Vox Sang 1972;23:528–536. antigen and gram-negative organisms. Transfusion 1979;19:663
57. Beck ML, Marsh WL, Pierce SR, et al: Auto-anti-Kpb associated (Abstr).
with weakened antigenicity in the Kell blood group system: A 82. Ellisor SS, Reid ME, O’Day T, et al: Autoantibodies mimicking
second example. Transfusion 1979;19:197–202. anti-Jkb plus anti-Jk3 associated with autoimmune hemolytic
256 Immune Hemolytic Anemias

anemia in a primipara who delivered an unaffected infant. Vox 109. Yokoyama M, Eith DT, Bowman M: The first example of auto-
Sang 1983;45:53–59. anti-Xga. Vox Sang 1967;12:138–139.
83. Grishaber JE, Cordle DG, Strauss RG: Development of alloanti- 110. Marsh WL, Brown PJ, DiNapoli J, et al: Anti-Wj: An autoanti-
Jka in a patient with hemolytic anemia due to autoanti-Jkb. J body that defines a high-incidence antigen modified by the
Clin Pathol 1992;98:542. In(Lu) gene. Transfusion 1983;23:128–130.
84. O’Day T: A second example of autoanti-Jk3. Transfusion 111. Fitzsimmons J, Caggiano V: Autoantibody to a high-frequency
1987;27:442. Lutheran antigen associated with immune hemolytic anemia
85. Issitt PD, Pavone BG, Frohlich JA, Mallory DM. Absence of and a hemolytic transfusion episode. Transfusion 1981;21:612
autoanti-Jk3 as a component of anti-dl. Transfusion 1980; (Abstr).
20:733–736. 112. Ward JM, Caggiano V: Clarification. Transfusion 1983;23:174.
86. Issitt PD, Pavone BG. Critical re-examination of the specificity 113. van Alphen L, Poole J, Overbeeke M: The Anton blood group
of auto-anti-Rh antibodies in patients with a positive direct antigen is the erythrocyte receptor for Haemophilus intuenzae.
antiglobulin test. Br J Haematol 1978;38:63. FEMS Microbiol Lett 1986;37:69.
87. van’t Veer MB, van Wieringen PMV, van Leeuwen I, et al: A 114. Denegri JF, Nanji AA, Sinclair M, Stillwell G: Autoimmune
negative direct antiglobulin test with strong IgG red cell hemolytic anemia due to immunoglobulin G with anti-Sdx
autoantibodies present in the serum of a patient with autoim- specificity. Acta Haematol 1983;69:19–22.
mune haemolytic anaemia. Br J Haematol 1981;49:383–386. 115. Sullivan CM, Kline WE, Rabin BI, et al: The first example of
88. Dickstein B, Kosanke J, Morris D, et al: Report of an autoanti- autoanti-Kx. Transfusion 1987;27:322–324.
body with mimicking allo anti-Fyb specificity. Transfusion 116. Marsh WL, DiNapoli J, Oyen R, et al: “New” autoantibody
1998;38:37S (Abstr). specificity in autoimmune hemolytic anemia defined with
89. Szymanski I, Roberts PL, Rosenfield RE: Anti-A autoantibody RBCs treated with 2-aminoethylisothiouronium bromide and
with severe intravascular hemolysis. N Engl J Med a dithiothreitol-papain solution. Transfusion 1985;
1976;294:995–996. 25:364–367.
90. Parker AC, Willis G, Urbaniak SJ, Innes EM: Autoimmune 117. Spielmann W: Spezifische autoantikörper bei hämolytischen
haemolytic anaemia with anti-A autoantibody. Br Med J Anämien. Klin Wschr 1956;34:248.
1978;1:26. 118. Meuli HC: Über blutgruppenspezifische antierythrocytäre
91. Govoni M, Turbiani C, Menini C, Tomasi P: Anti-A autoantibody Autoantikörper. Blut 1957;3:270.
associated with immune hemolytic anemia. Vox Sang 1991;61:75. 119. Fudenberg HH, Rosenfield RE, Wasserman LR: Unusual
92. McClelland WM, Bradley A, Morris TCM, et al: Auto-anti-B in specificity of auto-antibody in auto-immune hemolytic
a patient with acute leukaemia. Vox Sang 1981;41:231. disease. J Mt Sinai Hosp 1958;25:324.
93. Atichartakarn V, Chiewsilp P, Ratanasirivanich P, 120. Dunsford I, Stapleton RR: In vitro absorption of the Rhesus
Stabunswadgan S: Autoimmune hemolytic anemia due to antibody anti-D by Rhesus negative RBCs. Vox Sang
anti-B autoantibody. Vox Sang 1985;49:301–303. 1959;4:406.
94. Kuipers EJ, van Imhoff GW, Hazenberg CAM Smit J: Anti-H 121. Matuhasi T: Plasma protein and antibody fractions observed
IgM (Kappa) autoantibody mediated severe intravascular from the serological point of view. In Proceedings of 15th
haemolysis associated with malignant lymphoma. Br J General Assembly Japanese Medical Congress, Tokyo, Japan,
Haematol 1991;18:283–285. 1959:80.
95. Garratty G, Haffleigh B, Dalziel J, Petz LD: An IgG anti-IT 122. Ogata T, Matuhasi T: Problems of specific and cross reactivity
detected in a Caucasian American. Transfusion 1972;12:325. of blood group antibodies. In Proceedings of 8th Congress of
96. Booth PB, Jenkins WJ, Marsh WL: Anti-IT: A new antibody of International Society of Blood Transfusion, Tokyo, Japan,
the I blood-group system occurring in certain Melanesian sera. 1960:208.
Br J Haematol 1966;12:341–344. 123. Ogata T, Matuhasi T: Further observations on the problems of
97. Garratty G, Petz LD, Wallerstein RO, Fudenberg HH: specific and cross reactivity of blood group antibodies. Proc
Autoimmune hemolytic anemia in Hodgkin’s disease associ- 9th Congr Int Soc Blood Transf, Mexico 1962:528 (Karger,
ated with anti-I T. Transfusion 1974;14:226–231. Basel, New York 1964).
98. Garratty G, Petz LD, Wallerstein RO, Fudenberg HH: 124. Svardel JM, Yarbro J, Yunis EJ: Ogata phenomenon explaining
Development of the IT antigen. Transfusion 1974;14:630. the unusual specificity in eluates from Coombs-positive cells
99. Hafleigh EB, Wells RF, Grumet FC: Nonhemolytic IgG anti-IT. sensitized by autogenous anti-I. Vox Sang 1967;13:472.
Transfusion 1978;18:592–597. 125. Allen FH, Jr, Issitt PD, Degnan TJ, et al: Further observations
100. Levine AM, Thornton P, Forman SJ, et al: Positive Coombs test on the Matuhasi-Ogata phenomenon. Vox Sang 1969;16:47.
in Hodgkin’s disease: Significance and implications. Blood 126. Bove JR, Holburn AM, Mollison PL: Non-specific binding of IgG
1980;55:607–611. to antibody-coated RBCs (the “Matuhasi-Ogata Phenomenon”).
101. Freedman J, Newlands M, Johnson CA: Warm IgM anti-IT Immunology 1973;25:793–801.
causing autoimmune haemolytic anaemia. Vox Sang 127. Issitt PD, Pavone BG: Critical re-examination of the specificity
1977;32:135–112. of auto-anti-Rh antibodies in patients with a positive direct
102. Szatoky A, van der Hart M: An auto-antibody anti-Vel. Vox antiglobulin test. Br J Haematol 1978;38:63–70.
Sang 1971;20:376–377. 128. Issitt PD, Zellner DC, Rolih SD, Duckett JB: Autoantibodies
103. Herron R, Hyde RD, Hillier SJ: The second example of an anti- mimicking alloantibodies. Transfusion 1977;17:531–538.
Vel auto-antibody. Vox Sang 1979;36:179–181. 129. Issitt PD: Applied blood group serology, 3rd ed. Miami:
104. Ferrer Z, Cornwall S, Berger R, et al: A third example of Montgomery Scientific, 1985.
haemolytic auto-anti-Vel. Blood Transfus Immunohaematol 130. van’t Veer MB, van Wieringen PMV, van Leeuwen I, et al: A
1984;27:639. negative direct antiglobulin test with strong IgG red cell
105. Becton DL, Kinney TR: An infant girl with severe autoimmune autoantibodies present in the serum of a patient with auto-
hemolytic anemia: Apparent anti-Vel specificity. Vox Sang immune haemolytic anaemia. Br J Haematol 1982;52:537.
1986;51:108–111. 131. Issitt PD, Gruppo RA, Wilkinson SL, Issitt CH: Atypical pres-
106. McDowell MA, Stocker I, Nance S, Garratty G: Autoanti-Sc1 entation of acute phase, antibody-induced haemolytic
associated with autoimmune hemolytic anemia. Transfusion anaemia in an infant. Br J Haematol 1982;52:537–541.
1986;26:578 (Abstr). 132. Issitt PD, Wilkinson SL, Gruppo RA: Depression of Rh antigen
107. Owen I, Chowdhury V, Reid ME, et al: Autoimmune hemolytic expression in antibody-induced haemolytic anaemia. Br J
anemia associated with anti-Sc1. Transfusion 1992;32:173–176. Haematol 1983;53:688.
108. Peloquin P, Moulds M, Keenan J, Kennedy M: Anti-Sc3 as an 133. Vengelen-Tyler V, Mogck N: Two cases of “hrB-like” auto-
apparent autoantibody in two patients. Transfusion antibodies appearing as alloantibodies. Transfusion 1991;
1989;29:49S (Abstr). 31:254–256.
Specificity of Autoantibodies 257

134. Chown B, Kaita H, Lowen B, Lewis M: Transient production of 159. Kay MMB: Senescent cell antigen: A red cell aging antigen. In
anti-LW by LW-positive people. Transfusion 1971;11:220. Garratty G (ed): Red cell antigens and antibodies. Arlington,
135. Perkins HA, Mcllroy M, Swanson J, Kadin M: Transient LW- VA: American Association of Blood Banks, 1986:35.
negative red blood cells and anti-LW in a patient with 160. Gray LS, Kleeman JE, Masouredis SP: Differential binding of
Hodgkin’s disease. Vox Sang 1977;33:299–303. IgG anti-D and IgG autoantibodies to reticulocytes and red
136. Komatsu F, Kajiwara M: Transient depression of LWa antigen blood cells. Br J Haematol 1983;55:335–345.
with coincident production of anti-LWa repeated in relapses of 161. Branch DR, Shulman IA, Sy Siok Hian AL, Petz LD: Two dis-
malignant lymphoma. Transfus Med 1996;6:139. tinct categories of warm autoantibody reactivity with age-frac-
137. Mannessier L, Rouger P, Johnson CL, et al: Acquired loss of tionated RBCs. Blood 1984;63:177–180.
red-cell Wj antigen in a patient with Hodgkin’s disease. Vox 162. Herron R, Clark M, Smith DS: An autoantibody with activity
Sang 1986;50:240–244. dependent on red cell age in the serum of a patient with
138. van’t Veer MB, van Leeuwen 1, Haas FJLM, et al: Red-cell autoimmune haemolytic anaemia and a negative direct
auto-antibodies mimicking anti-Fyb specificity. Vox Sang antiglobulin test. Vox Sang 1987;52:71–74.
1984;47:88–91. 163. Hedge UM, Gordon-Smith EC, Worlledge SM: Reticulo-
139. Harris T: Two cases of autoantibodies that demonstrate mim- cytopenia and “absence” of red cell autoantibodies in immune
icking specificity in the Duffy blood group system. haemolytic anaemia. Br Med J 1977;2:1444–1447.
Immunohematology 1990;6:87. 164. Conley CL, Lippman SM, Ness PM, et al: Autoimmune
140. Dickstein B, Kosanke J, Morris D, et al: Report of an autoanti- hemolytic anemia with reticulocytopenia and erythroid
body with mimicking alloanti-Fyb specificity. Transfusion marrow. N Engl J Med 1982;306:281.
1998;38:37S (Abstr). 165. Hauke G, Fauser AA, Weber S, Maas D: Reticulocytopenia in
141. Issitt PD, Obarski G, Hartnett PL, Wren MR, Prewitt PL: severe autoimmune hemolytic anemia (AIHA) of the warm
Temporary suppression of Kidd system antigen expression antibody type. Blut 1983;46:321–327.
accompanied by transient production of anti-Jk3. Transfusion 166. Mangan KF, Besa EC, Shadduck RK, et al: Demonstration of
1990;30:46–50. two distinct antibodies in autoimmune hemolytic anemia with
142. Moulds M, Strohm P, McDowell MA, et al: Autoantibody mim- reticulocytopenia and red cell aplasia. Exp Hematol
icking alloantibody in the Colton blood group system. 1984;12:788–793.
Transfusion 1988;28:36S (Abstr). 166a. Davidyuk G, Goldsby RA, Dearden MT, Stec TC,
143. Williamson LM, Poole J, Redman C, et al: Transient loss of pro- Andrzejewski C: Autoantibodies to immature erythrocytes in
teins carrying Kell and Lutheran red cell antigens during con- patients with the anti-phospholipid antibody syndrome.
secutive relapses of autoimmune thrombocytopenia. Br J Transfusion 2002;42:103S (Abstr).
Haematol 1994;87:805–812. 167. Lutz HU, Wipf G: Naturally occurring autoantibodies to skele-
144. Leger R, Garratty G, Vengelen-Tyler V: Sequential suppression tal proteins from human red blood cells. J Immunol
of Lan and Lub antigens associated with mimicking anti-Lan 1982;128:1695–1699.
and anti-Lub. Transfusion 1994;34:69S (Abstr). 168. Wiener E, Hughes-Jones NC, Irish WT, Wickramasinghe SN:
145. Rand BP, Olson JD, Garratty G, et al: Coombs negative Elution of antispectrin antibodies from RBCs in homozygous
immune hemolytic anemia with anti-E occurring in the red thalassaemia. Clin Exp Immunol 1986;63:680–686.
blood cell eluate of an E-negative patient. Transfusion 169. Wakui H, Imai H, Kobayashi R, et al: Autoantibody against
1978;18:174–180. erythrocyte protein 4.1 in a patient with autoimmune
146. Rosenfield RE: Unpublished observations 1974, cited by Issitt hemolytic anemia. Blood 1988;72:408–412.
PD and Issitt CH in Applied blood group serology, 2nd ed., 170. Postoway N, Anstee DJ, Wortman M, Garratty G: A severe
Spectra Biologicals, Oxnard, 1975. transfusion reaction associated with anti-EnaTS in a patient
147. Rosenfield RE, Allen FH Jr, Swisher SN, et al: A review of Rh with an abnormal alpha-like red cell sialoglycoprotein.
serology and presentation of a new terminology. Transfusion Transfusion 1988;28:77–80.
1962;2:287. 171. Reid ME, Vengelen-Tyler V, Shulman I, Reynolds MV:
148. Lalezari P, Berens JA: Specificity and cross reactivity of cell- Immunochemical specificity of autoanti-Gerbich from two
bound antibodies. Human Blood groups. 5th Int. Convoc patients with autoimmune haemolytic anaemia and concomi-
Immunol, Buffalo, NY, 1976:44. tant alteration in the red cell membrane sialoglycoprotein. Br J
149. Issitt PD, Combs MR, Bumgarner DG, et al: Studies of anti- Haematol 1988;69:61–66.
bodies in the sera of patients who have made red cell autoan- 172. Gomperts ED, Metz J, Zail SS: Red cell membrane protein in
tibodies. Transfusion 1996;36:481–486. antibody-induced haemolytic anaemia. Br J Haematol
150. Beck ML, Dixon J, Oberman HA: Variation of specificity of 1973;25:421–428.
autoantibodies in autoimmune hemolytic anemia. Am J Clin 173. Kajii E, Ikemoto S, Ueki J, Miuras Y: Isolation of a peptide asso-
Pathol 1971;56:475–478. ciated with autoimmune haemolytic anaemia from red cell
151. Bird GWG, Wingham J: Changes in specificity of erythrocyte membranes. Clin Exp Immunol 1988;73:406–409.
autoagglutinins. Vox Sang 1972;22:364–365. 174. Hazeltine M, Rauch Y, Danoff D, et al: Antiphospholipid
152. Gerbal A, Homberg JC, Rochant H, et al: Les autoanticorps antibodies in systemic lupus erythematosus: Evidence of an
d’anemies hemolytiques acquises. I. Analyse de 2234 observa- association with positive Coombs’ and hypocomplementemia.
tions. Nouv Rev Frank Haematol 1968;8:155. J Rheumatol 1988;15:80–86.
153. Brendemoen OJ: A cold agglutinin specifically active against 175. Win N, Islam SIAM, Peterkin MA, Walker ID: Positive direct
stored RBCs. Acta Pathol Microbiol Scand 1952;31:574. antiglobulin test due to antiphospholipid antibodies in normal
154. Stratton F, Renton PH, Rawlinson VI: Serological difference healthy blood donors. Vox Sang 1997;72:182–184.
between old and young cells. Lancet 1960;1:1388. 176. Sthoeger Z, Sthoeger D, Green L, Geltner D: The role of anti-
155. Jenkins WJ, Marsh WL: Autoimmune haemolytic anaemia. cardiolipin autoantibodies in the pathogenesis of autoimmune
Lancet 1961;2:16. hemolytic anemia. Blood 1990;76(Suppl.):392a (Abstr).
156. Ozer FL, Chaplin H, Jr: Agglutination of stored erythrocytes 177. Cabral AR, Cabiedes J, Alarcon-Segovia D: Hemolytic anemia
by a human serum. Characterization of the serum factor and related to an IgM autoantibody to phosphatidylcholine that
erythrocyte changes. J Clin Invest 1963;42:1735. binds in vitro to stored and to bromelain-treated human
157. Easton JA, Priest CJ, Giles CM: An antibody against erythrocytes. J Autoimmun 1990;3:773–787.
stored blood associated with cirrhosis of the liver and false- 178. Arvieux J, Schweizer B, Roussel B, Colomb MG: Autoimmune
positive serological tests for syphilis. J Clin Pathol 1965;18:460. haemolytic anaemia due to anti-phospholipid antibodies. Vox
158. Arndt P, O’Hoski P, McBride J, Garratty G: Autoimmune Sang 1991;61:190–195.
hemolytic anemia associated with an antibody reacting prefer- 179. Guzmán J, Cabral AR, Cabiedes J, Pita-Ramirez L, Alarcón-
entially with “old” RBCs. Transfusion 1989;29:48S (Abstr). Segovia D: Antiphospholipid antibodies in patients with
258 Immune Hemolytic Anemias

idiopathic autoimmune haemolytic anaemia. Autoimmunity group A, B, H, Lea, Leb and precursor substances. J Exp Med
1994;18:51–56. 1972;135:1247–1258.
180. Lang B, Straub RH, Weber S, et al: Elevated anticardiolipin 205. Cooper AG, Brown MC: Serum i antigen: A new human blood
antibodies in autoimmune haemolytic anaemia irrespective group glycoprotein. Biochem Biophys Res Commun
of underlying systemic lupus erythematosus. Lupus 1997; 1973;55:297–304.
6:652–655. 206. Rosse WF, Lauf PK: Reaction of cold agglutinins with I antigen
181. Masouredis SP, Branks MJ, Garratty G, Victoria EJ: solubilized from human RBCs. Blood 1970;36:777–784.
Immunospecific red cell binding of iodine-125-labeled 207. Rosse WF, Sherwood JB: Cold-reacting antibodies: Differences
immunoglobulin G erythrocyte autoantibodies. J Lab Clin in the reaction of anti-I antibodies with adult and cord red
Med 1987;10:308–317. blood cells. Blood 1970;36:28–42.
182. Masouredis SP, Branks MJ, Victoria EJ: Antiidiotypic IgG cross 208. Gardas A, Koscielak J: I-active antigen of human erythrocyte
reactive with Rh alloantibodies in red cell autoimmunity. membrane. Vox Sang 1974;26:227–237.
Blood 1987;70:710–715. 209. Marcus DM, Kabat EA, Rosenfield RE: The action of enzymes
183. Garratty G: Specificity of autoantibodies reacting optimally at from clostridium tertium on the I antigenic determinant of
37°C. Immunohematology 1999;15:24–40. human erythrocytes. J Exp Med 1963;118:175.
184. Mino P: “La panemoagglutinina del sangue umane.” 210. Adamany AM, Kathan RH: Isolation of a tetrasaccharide
Policlinico, Sez. prat. 1924;31:1355. common to MM, NN and MN antigens. Biochem Biophys Res
185. Wiener AS, Unger LJ, Cohen L, Feldman J: Type-specific cold Commun 1969;37:171.
auto-antibodies as a cause of acquired hemolytic anemia and 211. Lisowska E: The degradation of M and N blood group glyco-
hemolytic transfusion reactions: Biologic test with bovine proteins and glycopeptides with alkaline borohydride. Eur J
RBCs. Ann Intern Med 1956;44:221. Biochem 1969;10:574–579.
186. Jenkins WJ, Marsh WL, Noades J, et al: The I antigen and anti- 212. Thomas DB, Winzler RJ: Structural studies on human erythro-
body. Vox Sang 1960;5:97. cyte glycoproteins: Alkali-labile oligosaccharides. J Biol Chem
187. Marsh WL, Jenkins WJ: Anti-i: A new cold antibody. Nature 1969;244:5943–5946.
1960;188:753. 213. Thomas DB, Winzler RJ: Structure of glycoproteins of human
188. Marsh WL: Anti-i: A cold antibody defining the Ii relationship erythrocytes: Alkali-stable oligosacharides. Biochem J
in human RBCs. Br J Haematol 1961;7:200. 1971;124:55–59.
189. Marsh WL, Nichols ME, Reid ME: The definition of two I 214. Dzierzkowa-Borodej W, Lisowska E, Seyfriedowa H: The
antigen components. Vox Sang 1971;20:209–217. activity of glycoproteins from erythrocytes and protein frac-
190. Dzierzkowa-Borodej W, Seyfried H, Nichols M, et al: The tions from human colostrum towards anti-I antibodies. Life Sci
recognition of water-soluble I blood group substance. Vox 1970;9:111–120.
Sang 1970;18:222–234. 215. Hamaguchi H, Cleve H: Solubilization of human erythrocyte
191. Marsh WL, Nichols ME, Allen FH: Inhibition of anti-I sera by membrane glycoproteins and separation of the MN glycopro-
human milk. Vox Sang 1970;18:149–154. tein from a glycoprotein with I, S and A activity. Biochem
192. Dzierzkowa-Borodej W, Seyfried H, Lisowska E: Serological Biophys Acta 1972;278:271–280.
classification of anti-I sera. Vox Sang 1975;28:110–121. 216. Lau FO, Rosse WF: The reactivity of red blood cell membrane
193. Lisowska E, Dzierzkowa-Borodej W, Seyfried H, et al: glycoprotein with cold-reacting antibodies. Clin Immunol
Reactions of erythrocyte glycoproteins and their degradation Immunopathol 1975;4:1.
products with various anti-I sera. Vox Sang 1975;28:122–132. 217. McGinniss MH, Binder RA, Kales AN, et al: Cold autoimmune
194. Giblett ER, Crookston MC: Agglutinability of RBCs by anti-i hemolytic anemia with autoanti-AI specificity: 51Chromium
in patients with thalssaemia major and other haematological survival studies. Immunohematology 1987;3:20–22.
disorders. Nature 1964;201:1138. 218. McGinniss MH: Auto-anti-AI in the serum of a patient with
195. Burnie K: Ii antigens and antibodies. Can J Med Technol fatal autoimmune hemolytic anemia. Immunohematology
1973;35:5–7. 1987;3:232–234.
196. van Loghen JJ, Peetom F, van der hart M, et al: Serological and 219. Postoway N, Garratty G, Guerra-Zevallos M: Transient
immunochemical studies in haemolytic anaemia with high- autoimmune hemolytic anemia (AIHA) associated with an
titre cold agglutinins. Vox Sang 1963;8:33. autoanti-IB in an A2B individual. In Book of Abstracts from
197. Worlledge SM, Dacie JV: Haemolytic and other anaemias in the ISBT/AABB Joint Congress. Arlington, VA: American
infectious mononucleosis. In: Carter RL, Penman, (eds): Association of Blood Banks, 1990:27 (Abstr).
Infectious mononucleosis. Oxford: Blackwell Scientific 220. Roelcke D: Sialic acid-dependent red blood cell antigens. In
Publications, 1969. Garratty G (ed): Immunobiology of transfusion medicine.
198. Wilkinson LS, Petz LD, Garratty G: Reappraisal of the role of New York: Dekker, 1994:69–95.
anti-i in haemolytic anaemia in infectious mononucleosis. Br J 221. Marsh WL, Jenkins WJ: Anti-Sp1: The recognition of a new cold
Haematol 1973;25:715–722. autoantibody. Vox Sang 1968;15:177–186.
199. Tegoli J, Harris JP, Issitt PD, et al: Anti-IB, an expected “new” 222. Roelcke D: A new serological specificity in cold antibodies of
antibody detecting a joint product of the I and B genes. Vox high titre: Anti-HD. Vox Sang 1969;16:76–79.
Sang 1967;13:144–157. 223. Roelcke D, Uhlenbruck G, Bauer K: A heterogeneity of the HD-
200. Morel P, Garratty G, Willbanks E: Another example of anti-IB. receptor, demonstrable by HD-cold antibodies: HD1/HD2.
Vox Sang 1975;29:231–233. Scand J Haematol 1969;6:280.
201. Feizi T, Kabat EA, Vicari G, et al: Immunochemical studies on 224. Roelcke D, Uhlenbruck G: Letter to the editor. Vox Sang
blood groups. XLVII. The I antigen complex-precursors in the 1970;18:478–479.
A, B, H, Lea and Leb blood group system-hemagglutinin-inhi- 225. Roelcke D. Cold agglutinin: Antibodies and antigens. Clin
bition studies. J Exp Med 1971;133:39–52. Immunol Immunopathol 1974;2:266.
202. Vicari G, Kabat EA: Immunochemical studies on blood 226. Roelcke D, Anstee DJ, Jungfer H, Nutzenadel W, Webb AJ:
groups. XLV. Structures and activitives of oligosaccharides IgG-type cold agglutinins in children and corresponding anti-
produced by alkaline degradation of a blood group substance gens: Detection of a new Pr antigen: Pra. Vox Sang 1971;20:
lacking A, B, H, Lea and Leb specificities. Biochemistry 218–229.
1970;9:3414. 227. Roelcke D, Ebert W, Geisen HP: Anti-Pr3: serological and
203. Watkins WM, Morgan WTJ: Possible genetical pathways of immunochemical identification of a new anti-Pr subspecificity.
blood group mucopolysaccharides. Vox Sang 1959;4:97. Vox Sang 1976;30:122–133.
204. Feizi T, Kabat EA: Immunochemical studies on blood groups. 228. Dellagi K, Brouet JC, Schenmetzler C, Praloran V: Chronic
LIV. Classification of anti-I and anti-i sera into groups based on hemolytic anemia due to a monoclonal IgG cold agglutinin
reactivity patterns with various antigens related to the blood with anti-Pr specificity. Blood 1981;57:189–191.
Specificity of Autoantibodies 259

229. Roelcke D, Anstee DJ, Jungfer H, Nutzenadel W, Webb AJ: anti-Rx (previously anti-Sdx). Clin Lab Haematol 1988;
IgG-type cold agglutinins in children and corresponding anti- 10:105–108.
gens: Detection of a new Pr antigen: Pra. Vox Sang 1971; 252. Salama A, Pralle H, Mueller-Eckhardt C: A new red blood cell
120:218–229. cold autoantibody (anti-Me). Vox Sang 1985;49:277–284.
230. Meytes D, Adler M, Viraq I, Feigl D, Levene C: High-dose 253. Götsche B, Salama A, Mueller-Eckhardt C: Autoimmune
methylprednisolone in acute immune cold hemolysis. N Engl hemolytic anemia caused by a cold agglutinin with a new
J Med 1985;312:318. specificity (anti-Ju). Transfusion 1990;30:261–262.
231. Northoff H, Martin A, Roelcke D: An IgG-monotypic anti-Pr1h 254. Perrault R: “Cold” IgG autologous anti-LW. Vox Sang
associated with fresh varicella infection. Eur J Haematol 1973;24:150–164.
19987;38:85–88. 255. Longster GH, Johnson E: IgM anti-D as auto-antibody in a case
232. Curtis BR, Lamon J, Roelcke D, Chaplin H: Life-threatening, of “cold” autoimmune haemolytic anaemia. Vox Sang
antiglobulin test-negative, acute autoimmune hemolytic 1988;54:174–176.
anemia due to a non-complement-activating IgG1 cold anti- 256. Beaumont JL, Lorenzelli L, Delplanque B, et al: A new serum
body with Pra specificity. Transfusion 1990;30:838–843. lipoprotein associated erythrocyte antigen which reacts with a
233. Kadota Y, Fujinami S, Tagawa Y, et al: Haemolytic anaemia monoclonal IgM. Vox Sang 1976;30:36–49.
caused by anti-Pra following rubella infection. Transfus Med 257. Linder E, Edgington TS: Immunobiology of the autoantibody
1993;3:207. response. 11. The lipoprotein-associated soluble HB erythro-
234. Ramos RR, Curtis BR, Sadler JE, Eby CS, Chaplin H: Refractory cyte autoantigen of NZB mice. J Immunol 1973;110:53–62.
immune hemolytic anemia with a high thermal amplitude, 258. Lightwood AM, Scott GL: Autoimmune haemolytic anaemia
low affinity IgG anti-Pra cold autoantibody. Autoimmunity due to red cell antibodies of different specificities in a patient
1992;12:149–154. with chronic hepatitis. Vox Sang 1973;24:331–336.
235. Angevine CD, Andersen BR, Barnett EV: A cold agglutinin of 259. Cazzola M, Barosi G, Ascari E: Cold haemagglutinin disease
the IgA class. J Immunol 1966;96:578–586. with severe anaemia, reticulocytopenia and erythroid bone
236. Roelcke D, Dorow W: Besonderheiten der Reaktionswerte marrow. Scand J Haematol 1983;30:25.
eines mit Plasmocytom-?A-Paraprotein identischen 260. Kosmin M, Tarantolo S, Arndt P, Garratty G: Cold agglutinin
Kalteagglutinins. Klin Wschr 1968;46:126–131. syndrome associated with an autoagglutinin reacting prefer-
237. Garratty G, Petz LD, Brodsky I, et al: An IgA high-titer cold entially with young RBCs (Abstr). In Book of Abstracts from
agglutinin with an unusual blood group specificity within the the ISBT/AABB Joint Congress. Arlington, VA: American
Pr complex. Vox Sang 1973;25:32–38. Association of Blood Banks, 1990:86.
238. Tonthat H, Rochant H, Henry A, et al: A new case of mono- 261. Levine P, Celano MJ, Falkowski F: The specificity of the anti-
clonal IgA Kappa cold agglutinin with anti-Pr1d specificity in body in paroxysmal cold hemoglobinuria (PCH). Transfusion
a patient with persistent HB antigen cirrohosis. Vox Sang 1963;3:278.
1976;30:464–468. 262. Worlledge SM, Rousso C: Studies on the serology of paroxys-
239. Roelcke D: Specificity of IgA cold agglutinins: Anti-Pr1. Eur J mal cold haemoglobinuria (PCH), with special reference to its
Immunol 1973;3:206–212. relationship with the P blood group system. Vox Sang
240. Gergely J, Wang AC, Fudenberg HH: Chemical analyses of 1965;10:293.
variable regions of heavy and light chains of cold agglutinins. 263. Knapp T: The laboratory investigation of three cases of paro-
Vox Sang 1973;24:432–440. xysmal cold haemoglobinuria. Can J Med Tech 1964;26:172.
241. Wang AC, Fudenberg HH, Wells JV, et al: A new subgroup of 264. Vogel JM, Hellman M, Moloshok RE: Paroxysmal cold hemo-
the kappa chain variable region associated with anti-Pr cold globinuria of nonsyphilitic etiology in two children. J Pediatr
agglutinins. Nat New Biol 1973;243:126–128. 1972;81:974–977.
242. Sokol RJ, Booker DJ, Stamps R, et al: Autoimmune haemolysis 265. Ries CA, Garratty G, Petz LD, et al: Paroxysmal cold hemoglo-
and red cell autoantibodies with ABO blood group specificity. binuria: report of a case with an exceptionally high thermal
Haematologia 1995;26:121–129. range Donath-Landsteiner antibody. Blood 1971;38:491–499.
243. Postoway N, Garratty G, Guerra-Zevallos M: Transient 266. Bird GWG, Wingham J, Martin AJ, et al: Idiopathic non-
autoimmune hemolytic anemia (AIHA) associated with an syphlitic paroxysmal cold haemoglobinuria in children. J Clin
autoanti-IB in an A2B individual. In: Book of Abstracts from Pathol 1976;29:215–218.
the ISBT/AABB Joint Congress, 1990:27 (Abstr). 267. Johnsen HE, Brostrøm K, Madsen M: Paroxysmal cold haemo-
244. Arndt P, Do J, Garratty G, et al: A high titer, high thermal globinuria in children: 3 cases encountered within a period of
amplitude autoanti-B associated with acrocyanosis but not 7 months. Scand J Haematol 1978;20:413.
obvious hemolytic anemia. Transfusion 2003;43:1133–1137 268. von dem Borne AEG Kr, Mol JJ, Joustra-Maas N, et al:
(Abstr). Autoimmune haemolytic anaemia with monoclonal IgM (K)
245. Sangster JM, Kenwright MG, Walker MP, Pembroke AC: Anti anti-P cold autohaemolysins. Br J Haematol 1982;50:345–350.
blood group-M autoantibodies with livedo reticularis, 269. Ramos RR, Curtis BR, Eby CS, Ratkin GA, Chaplin H: Fatal
Raynaud’s phenomenon, and anaemia. J Clin Pathol 1979; outcome in a patient with autoimmune hemolytic anemia
32:154–157. associated with an IgM bithermic anti-ITP. Transfusion
246. Chapman J, Murphy MF, Waters AH: Chronic cold haemag- 1994;34:427–431.
glutinin disease due to an anti-M-like autoantibody. Vox Sang 270. Engelfriet CP, Beckers D, von dem Borne AEG Kr, et al:
1982;42:272–277. Haemolysins probably recognizing the antigen p. Vox Sang
247. Bowman HS, Marsh WL, Schumacher HR, et al: Autoanti-N 1971;23:176.
immunohemolytic anemia in infectious mononucleosis. Am J 271. Weiner W: The specificity of the antibodies in acquired
Clin Pathol 1974;61:465–472. haemolytic anaemias. In: Proceedings of the Joint Meeting
248. Marsh WL, Johnson CL, Oyen R, Nichols ME, et al: Anti-Sdx : of the 10th Congress of the International Society of
A “new” auto-agglutinin related to the Sda blood group. Haematology/l0th Congress of the International Society of
Transfusion 1980;20:1. Blood Transfusion 1964:24.
249. Bass LS, Rao AH, Goldstein J, Marsh WL: The Sdx antigen and 272. Shirey RS, Park K, Ness PM, et al: An anti-i biphasic hemolysin
antibody: biochemical studies on the inhibitory property of in chronic paroxysmal cold hemoglobinuria. Transfusion
human urine. Vox Sang 1983;44:191–196. 1986;26:62–64.
250. Marsh WL, Johnson CL, DiNapoli J, et al: Immune hemolytic 273. Judd WJ, Wilkinson SL, Issitt PD, et al: Donath-Landsteiner
anemia caused by autoanti-Sdx: A report on six cases (Abstr). hemolytic anemia due to an anti-Pr-like biphasic hemolysin.
Transfusion 1980;20:647. Transfuison 1986;26:423–425.
251. O’Brien DA, Mullahy DE, Garvey MA, Jackson JF: Cold 274. Bell CA, Zwicker H, Rosenbaum DL: Paroxysmal cold
autoimmune haemolytic anaemia in a 3-year-old infant due to hemoglobinuria (PCH) following mycoplasma infection:
260 Immune Hemolytic Anemias

Anti-I specificity of the biphasic hemolysin. Transfusion 276. Green ES, Devenish A, Bradshaw HH, et al: Chronic hae-
1973;13:138–141. molysis caused by an unusual Donath-Landsteiner-like (DL)
275. Nakamura H, Watanabe T, Hayashi T, et al: Donath-Landsteiner antibody. Transfus Med 1990;1:15 (S1).
antibody of the IgM class with anti-I specificity and possible 277. Sokol RJ, Booker DJ, Stamps R: Paroxysmal cold haemoglobi-
efficacy of azathiprine therapy in paroxysmal cold hemoglobin- nuria: A clinico-pathological study of patients with a positive
uria: A case report. Jpn J Clin Hematol 1990;31:1548–1552. Donath-Landsteiner test. Hematology 1999;4:137–164.
C H A P T E R 8

Drug-Induced Immune
Hemolytic Anemia

Drug-induced immune hemo- was positive, and the patient’s serum was shown to
lytic anemia (DIIHA) is rare. It react with allogeneic RBCs only when the drug was
is about one tenth as common present. The drug was stopped, and the patient’s
as AIHA.1 The purported inci- hemoglobin returned to normal in 20 days; the sero-
dence of AIHA in the general logy became negative after about 60 days.
population is approximately 1 In 1967, Dausset and Contu5 reviewed the literature
in 80,000.2 Thus, DIIHA would on drug-induced IHA and found only 34 published
occur in only about 1 in 1 cases, which were due to 15 drugs. In 1969, Worlledge6
million individuals. In San could add six extra cases, but no further drugs, to the
Francisco, we were referred list. By 1980, we had found reports of approximately
43 cases over a 10-year period (1970–1980).1 In Los 33 drugs as causes of IHA.1 By 1989, the list had grown
Angeles, we have had a similar experience, with 83 to more than 70 drugs that were reasonably well-
cases referred over a 20-year period (1983–2002). documented to cause IHA; many additional but less
well-documented examples also have appeared in the
literature.7 As of 2003, there are about 100 drugs that
INTRODUCTION have been incriminated in DIIHA (a few of the
included drugs have caused positive DATs without
Drugs were first suspected as a cause of immune obvious IHA) (Table 8-1).
hemolytic anemia (IHA) in 1953, when Snapper and There are many more drugs besides those that appear
coworkers3 described a patient who developed pancy- in Table 8-1 that have been reported to cause DIIHA.
topenia with hemolytic anemia, associated with a Unfortunately, many of the reports have no serologic
positive direct antiglobulin test (DAT), following data supporting that the etiology was immune-related.
ingestion of mephenytoin (Mesantoin). Harris4 was the George and colleagues8 came to similar conclusions
first to carefully document the history and serology after studying the literature concerning drug-induced
of a case of IHA due to a drug. The drug, apronalide thrombocytopenia. They determined that only 48% of
(Stibophen), was used to treat schistosomiasis. The 247 patient case reports supported a definite or probable
patient had received the drug 10 years previously with cause and role for the reported drug.
no problems. During a second episode of infection, Often, the published reports of DIIHA are a clini-
he developed acute intravascular hemolysis. The DAT cal summary of a patient developing hemolytic

261
262 Immune Hemolytic Anemias

TABLE 8-1. DRUGS CAUSING IHA OR POSITIVE DAT LISTED BY THERAPEUTIC CATEGORY

Antimicrobial

amphotericin B cefotetan erythromycin p-aminosalicylic acid sulfonamides


ampicillin cefoxitin fluoroquinolone penicillin G teicoplanin
carbenicillin ceftazidime isoniazid piperacillin temafloxacin
cefamandole ceftizoxime latamoxef quinine tetracycline
cefazolin ceftriaxone mefloquine rifampicin
cefixime cephalexin methicillin stibophen
cefotaxime cephalordine nafcillin streptomycin
β-Lactamase inhibitors
clavulanate potassium (e.g., in Timentin)
sulbactam sodium (e.g., in Unasyn)
tazobactam (e.g., in Zosyn)
Nonsteroidal anti-inflammatory
aceclofenac diclofenac ibuprofen propyphenazone
acetaminophen/paracetamol dipyrone indene derivatives (e.g., sulindac) suprofen
aminopyrine/pyramidon etodolac mefenamic acid tolmetin
apazone/azapropazone fenoprofen phenacetin zomepirac
Antineoplastic
carboplatin fluorouracil (5-FU) interleukin-2
cisplatin 9-hydroxy-methyl-ellipticinium 6-mercaptopurine
cladrabine teniposide methotrexate (also anti-rheumatic)
diglycoaldehyde (INOX) interferon (also anti-viral) oxaliplatin
fludarabine melphalan rituximab
Diuretic Anti-hypertensive
butizide (and antihypertensive) butizide (and diuretic)
furosemide hydralazine
hydrochlorothiazide methyldopa
triamterene
Antidiabetic Antiarrhythmic
insulin procainamide
sulfonylurea derivatives (e.g., chloropropamide) quinidine (also anti-malarial)
tolbutamide
Analgesic Sedative, hypnotic
glafenine apronalide
methadone carbromal
Miscellaneous
antazoline (antihistamine) fenfluramine (anorexic) phenytoin (anticonvulsant)
carbimazole (antithyroid) fluorescein (injectable dye) probenicid (urisocuric)
catergen/cyanidanol (antidiarrheal) levodopa (antiparkinsonian) ranitidine (antiulcerative)
chlorinated hydrocarbons (insecticides) mephenytoin (anticonvulsant) sodium pentothal/thiopental (anesthetic)
chlordiazepoxide (anxiety disorders) methotrexate (antirheumatic and anti- trimellitic anhydride (used in preparation of
chlorpromazine (antiemetic, antipsychotic) neoplastic) resins, dyes, adhesives, etc.)
cyclofenil (gonad stimulant) methysergide (antimigraine)
diethylstilbestrol (estrogen) nomifensine (antidepressant)

anemia (HA) after starting on a particular drug and able to show the presence of a drug-dependent anti-
the HA resolving after the drug was stopped. Even if body in an eluate from the patient’s DAT-positive
the patient had a positive DAT, these findings are not RBCs. The presence of a drug-dependent antibody in
sufficient to prove that the drug caused IHA. One the patient’s serum is supportive but not conclusive
must prove that there is a drug-dependent antibody evidence, as patients without hemolytic anemia and
(i.e., drug was required to be present to demonstrate healthy individuals can have drug-dependent
the antibody’s presence in vitro), or that a drug- antibodies (e.g., penicillin antibodies) in their sera.
independent antibody (i.e., autoantibody) was In addition, sometimes a positive indirect antiglo-
induced by the drug. This is very difficult to prove, bulin test (IAT) can be obtained because RBCs coated
as will be discussed later in this chapter. It is prefer- with some drugs (e.g., cephalosporins) take up
Drug-Induced Immune Hemolytic Anemia 263

proteins nonimmunologically (i.e., no antibody binding between the haptenic group and the carrier
involved). molecule. In its original meaning as used by
There are many reports in the literature in which the Landsteiner,11 however, the term hapten (from the
patient appears to produce RBC autoantibodies (i.e., Greek word for binding) did not imply a notion of size
drug-independent antibodies) following therapy with for the haptenic molecule and did not imply covalent
a particular drug. As such antibodies do not require binding between the haptenic group and the carrier.
drug for their in vitro reactions with RBCs, it is impos- Although the classical concepts of an immune response
sible to prove by serology that they were induced by a to low MW substances involves covalent bonding,
drug. The only way to prove that the drug caused the exceptions have been described. The Forssman hapten,
AIHA is to stop the drug, observe the cessation of for example, becomes immunogenic upon binding to
hemolysis and the resolution of serologic findings, an immunogenic carrier through noncovalent bonds.
and then start the drug again and show that the Nucleic acids and oligonucleotides, which are acidic
autoantibody reappears. Unfortunately, the hemolytic polymers, can be immunogenic if complexed to basic
anemia might also reappear, so this approach is rarely protein carriers (e.g., methylated bovine albumin);
used; however, the approach was used in the 1960s to thus, it would appear that multiple salt linkages might
prove that methyldopa caused AIHA.9 Petz and be sufficient to allow a haptenic response.15-17
coworkers10 published a report that illustrates the The immune response to drugs is affected by the
dangers of assuming that a drug is the cause of a coin- nature of the carrier molecule, the degree of conjuga-
cidental appearance of AIHA. They described two tion (epitope density), and the nature of the chemical
patients who developed HA following administration bond. Administration of benzylpenicillin (BP) to rats
of cimetidine and resolution of their hemolytic anemia at doses up to 2.7 mmol/kg (1 g/kg body weight)
and serology following cessation of drug therapy. does not evoke antibodies to the BP determinant, but
Both patients were started on the drug again and fol-
lowed up (for up to 24 months in the case of one
patient), with no recurrence of hemolytic anemia or
autoantibodies. This lack of recurrence almost cer- Chemical group
or structure Name
tainly proved that the drug had nothing to do with the
symptoms and that the start and finish of hemolysis sulfhydryl
-SH
paralleling the beginning and end of drug therapy disulfide
-S-S-
was coincidence. Unfortunately, it is very hard to -NH-NH2 hydrozide
perform such important confirmatory in vivo experi- -N=N- diozo
ments; indeed, it could be dangerous in a patient -N(CH2CH2CI)2 or -S(CH2CH2CI)2 mustard derivatives
whose initial symptoms included acute intravascular O acetoxymethylene
hemolysis rather than only a positive DAT or milder
=

- CH2- O - C- CH3
extravascular RBC destruction.
In this chapter, we have attempted to restrict our- - CH=CH2 styrene derivatives
selves to a discussion of drugs for which there is rea-
sonable evidence in the literature that the drug was
responsible for immune hemolytic anemia. We indicate O= =O quinones
whether some reports are suggestive but not conclusive.
-CH CH
β -lociom
THE IMMUNE RESPONSE TO DRUGS C N-
=

Drugs are small-molecular-weight (MW) substances O epoxy ring


(e.g., penicillin has an MW of 300 kD). Because of their
size, it is usually assumed that drugs act as haptens to R1 R2
evoke an immune response. If a hapten forms stable R3 substituted aromatic ring
bonds with macromolecules (e.g., protein), the result- R4
ing drug-protein conjugate could be immunogenic.11-13 R5
Haptens that form stable bonds with proteins do so Cl Cl
with nucleophilic groups on proteins. Figure 8-1 shows
Cl
chemical groups or structures that might bind firmly to CCl2 chlorinated hydrocarbons
protein (e.g., RBC membrane). Nucleophilic groups on Cl
proteins include lysyl and cysteinyl residues and the Cl
imidazole and phenol groups present in histidine and
tyrosine, respectively.14 De Weck15 points out that since FIGURE 8-1. Chemical groups or structures that may bind covalently
to soluble or erythrocyte membrane proteins under physiological condi-
Landsteiner’s original observations,11 many immuno- tions. (From Petz LD, Branch DR: Drug-induced immune hemolytic
logists have come to consider haptens as substances of anemia. In Chaplin H Jr (ed): Immune hemolytic anemias. New York:
low MW and the word conjugate as implying covalent Churchill Livingstone, 1985:47–94.)
264 Immune Hemolytic Anemias

formed might not react with the parent drug in vitro


(see the discussion later in this chapter).
Most of the drugs listed in Table 8-1 probably do
not form stable (e.g., covalent) bonds with proteins.
This is simply illustrated by the fact that RBCs
cannot be coated in vitro with most of the drugs (i.e.,
they are removed simply by washing the RBCs after
incubation in the drug), presumably because the
drugs do not combine well with membrane proteins.
A few of the drugs (e.g., quinidine) have been
studied by more sophisticated methods and have
been proven to have a low affinity for protein (see
FIGURE 8-2. Portions of the van der Waal’s outline of the
p-azobenzoate group coupled to protein that are reactive with various later discussion). Penicillin—and, probably, closely
antibodies, A-G = antibody combining sites of different antibody related drugs such as some of the cephalosporins—
molecules. (From Kitagawa M, Yagi Y, Pressman D: The heterogeneity do form stable covalent bonds with protein (e.g., on
of combining sites of antibodies as determined by specific immuno- RBC membranes). Penicillins contain a structure that
adsorbents. II. Comparison of elution patterns obtained with
p-azobenzoate antibodies by different kinds of immunoadsorbent and
reacts with nucleophilic amino, hydroxy, mercapto,
eluting hapten. J Immunol 1965;95:455. Copyright 1965, The and histidine groups. A number of sites on the peni-
American Association of Immunologists, Inc.) cillin molecule are open to nucleophilic attack and,
therefore, a number of antigenic determinants can be
a 1 million-fold lower dose of BP conjugated to a foreign formed. The major haptenic determinant is the
protein is readily immunogenic.18 Kristoffersen and benzlpenicilloyl (BPO) group.21-28 Cephalosporins
associates19 reported that a conjugate containing 11 peni- are related to penicillins in that both contain a β-
cillyl residues per each bovine serum albumin molecule lactam ring; cephalosporins have a dihydrothiazine
induced a significant antibody response in mice after a ring, whereas penicillins have a thiazolidine ring
single injection. When the ratio of penicillyl residues per (Fig. 8-3).29 Cephalosporins probably form a
bovine serum albumin is less than one, as might be cephalosporyl group equivalent to the penicilloyl
anticipated during therapeutic administration, there is group, but there is no conclusive evidence for this.
no detectable response, even after three injections. The chemistry of hapten formation by cephalo-
The haptenic group formed by small reactive sub- sporins has been hindered by the fact that a number
stances generally represents only a portion of the of unstable intermediates are formed during aminol-
antigen determinant, which also could encompass ysis of cephaloporins.13
part of the macromolecular carrier.11,20 A good A few other drugs have been shown to combine in
example of this appears in Figure 8-2, which shows vitro with RBC membranes efficiently enough to with-
the heterogeneity of the combining sites of antibodies stand multiple washes, but the exact mechanism of cell
to p-azobenzoate coupled to protein.20 Drugs that are binding is unknown. Because of the efficient binding,
hydrolysed rapidly cannot function as effective these drugs can be used to prepare drug-coated RBCs
haptens.13 The chemical structure of the antigenic to detect antibodies to such drugs. Table 8-2 lists these
determinant comprising the drug (or its metabolite) drugs.
and its carrier component might be quite different As can be seen from Tables 8-1 and 8-2, most drugs
from that of the parent drug, and the antibodies causing DIIHA do not bind efficiently to proteins (e.g.,

PENICILLINS

S CH3 S CH3
R CO NH CH CH C R1 CO NH CH CH C
CH3 CH3
O C N CH O C NH CH
COOH COOH
FIGURE 8-3. General structure of penicillins and
NH
cephalosporins. Substitutions at the R1 and R2 posi-
tions create the various commercially available anti-
biotics. The breakdown products that most commonly Protein
form protein conjugates are indicated to the right of
CEPHALOSPORINS
the arrows. (From Garratty G: Immune cytopenia asso-
ciated with antibiotics. Transfus Med Rev
S S
1993;VII:255–267.) R1 CO NH CH CH CH2 R1 CO NH CH CH CH2

O C N C R2 O C NH C R2
C C
NH
COOH COOH

Protein
Drug-Induced Immune Hemolytic Anemia 265

RBC membranes), so how do they act as immunogens? lyzed away, and the platelets were separated from the
The most popular hypotheses are discussed next. antibody; the antibody could be made to combine
with platelets again if further drug was added. This
The Hapten Hypothesis seemed to be clear evidence that the drug acted as a
link between the platelets and the antibody and
Ackroyd30-34 observed that, in cases of drug-induced strongly supported the concept that the drug acted as
thrombocytopenia, a factor was often readily demon- a hapten.
strated in the patient’s serum that caused agglutina- In these papers, Ackroyd criticized some aspects of
tion of the patient’s own platelets and of compatible his own hypothesis.30-34 He agreed that some elements
normal platelets in vitro in the presence of the drug of the classical definition of a hapten were not met.
but not in its absence. The quantity of the drug used The drug was not covalently bound to the platelets,
in vitro was not critical, and the reaction was not and the antibody could not be inhibited by adding
inhibited by large concentrations of the drug. In the high concentrations of the drug to the patient’s serum.
presence of complement, this factor also caused On the other hand, Ackroyd pointed out that
platelet lysis. Ackroyd determined that this factor was Landsteiner11 had shown that hapten-antibody com-
found in the gamma globulin fraction of the patient’s plexes had a considerable tendency to dissociate, and
serum. The gamma globulin nature of the factor, the that in consequence, very high concentrations of drug
fact that it caused agglutination of platelets in the were sometimes necessary to inhibit the antibody. He
absence of complement and caused platelet lysis in also pointed out that a thousand molecules in solution
the presence of complement, all led Ackroyd to con- might be required to inhibit the action of a single
clude that it was an antibody. artificially conjugated hapten molecule. Ackroyd32
It was clear from these simple experiments that the suggested that perhaps the simplest explanation of
antigen cannot be the platelet alone, as platelets were the high concentrations of the drug that are required
not agglutinated by the antibody except in the presence for in vitro and in vivo experiments is that high con-
of the drug. Ackroyd proposed, therefore, that centrations are needed to force the drug into combi-
the antigen was formed by union of the drug with the nation with the platelets to form the antigen. To
platelet, and suggested that the drug modified the explain why these concentrations do not inhibit the
platelet sufficiently that it became “autoantigenic”; he reaction, it seemed necessary further to postulate that
suggested that the drug acted as a hapten conferring the antibody was incapable of reacting with the drug
antigenic properties on the platelet. This implied that alone and could react only with the drug-platelet con-
the drug, on the surface of the platelet, formed the jugate. With reference to the binding of the drug to
determinant group with which the antibody combined, protein (e.g., membrane protein), as mentioned pre-
a mechanism that he referred to as a partial autoim- viously, it is now well established that covalent
munoreaction. Evidence in support of this hypothesis bonding to protein is not necessary for immunogenic-
was provided by the following observations. ity of low MW chemicals. In addition, Park and col-
First, if platelets were incubated with the serum of a leagues13 have pointed out that the ability of the drug
sensitized patient, in the presence of the drug but in to conjugate to proteins in vitro does not necessarily
the absence of complement, and the platelets were reflect the potential of the compound to form haptens
then washed, both antibody and drug were removed in vivo, as numerous alternative biotransformations
from the platelets. Second, if the platelets in the above might be available that will either preclude reactive
experiment were washed, not in saline, but in a satur- metabolite formation or deactivate an electrophilic
ated solution of the drug in saline, the antibody species once it has been formed. A number of drugs
remained in contact with the platelets. If the platelets that are associated with hypersensitivity reactions of
were now dialyzed against saline, the drug was dia- one form or another have the potential to form
haptens as a consequence of enzyme-catalyzed bio-
transformations in vivo.12 Park and colleagues13 sug-
gested that interindividual variation in ability and
capacity to perform a variety of biotransformations
TABLE 8-2. DRUGS THAT HAVE BEEN SHOWN are important determinants of drug-protein conjuga-
TO COMBINE WITH THE RBC MEMBRANE tion in vivo. They also emphasized that much of the
IN VITRO EFFICIENTLY ENOUGH TO WITHSTAND information concerning the immunogenicity of
MULTIPLE WASHES various substances has been obtained in animal
models designed to be especially sensitive to small
azapropazone cianidanol
carbimazole cisplatin
doses of putative immunogen, and that adjuvants are
carboplatin erythromycin often used.13 Caution, therefore, should be exercised
carbromal isoniazid when extrapolating the results of such studies to
cefotaxime 6-mercaptopurine humans.
cefotetan penicillins (most) Ackroyd’s hypothesis30-34 regarding the pathoge-
cefoxitin streptomycin
cephalothin tolbutamide nesis of drug-induced cytolytic antibodies is often
referred to as the “hapten mechanism.” This is a
266 Immune Hemolytic Anemias

rather unfortunate choice of terms, as, regardless of determinants. Most of the antibody activity (about
the mechanism by which drug antibodies are devel- 95%) is directed to a breakdown product of the parent
oped, a prerequisite to becoming immunogenic is the molecule having so-called penicilloyl determinants. In
formation of a haptenic conjugate. Thus, the “hapten addition to the major penicilloyl immunogenic deter-
mechanism,” the “immune complex mechanism,” and minant, other minor determinants have been defined.
the “drug adsorption mechanism” must all require The major breakdown product of the penicillins is peni-
conjugation of drug (hapten) with a macromolecule to cilloic acid, derived by hydrolysis of the β-lactam ring.
form the epitope that originally was recognized as Penicilloic acid does not have penicilloyl determinants,
foreign (i.e., the immunogen). Ackroyd’s work sug- but some skin-sensitizing antibodies appear to be
gested that the drug forms a labile combination with directed to this molecule.38 Skin-sensitizing antibodies
cellular proteins and that this drug-cell combination is also have been demonstrated to react with several
potentially antigenic, with the drug acting as a degradation products of penicilloic acid.38
hapten. The drug and cellular membrane proteins are Haptenization is thought to occur by aminolysis of
essential for immune stimulation and for antibody the β-lactam ring of penicillins by protein lysine
reactivity. residues. In addition, opening of the thiazolidine ring
exposes a thiol group, which can form mixed disulfide
bonds with cysteine or cystine residues on protein (see
The Immune Response to Penicillin Fig. 8-3). It is not easy to explain the formation of peni-
In 1958, Ley and associates35 were the first to demon- cilloyl conjugates in vivo, either by direct aminolysis
strate that humans could make penicillin antibodies. or by isomerization of penicillin to penicillinic acid,
During routine blood banking procedures, the serum and subsequent reaction with amino groups on pro-
of a prospective transfusion recipient was found to teins. Both of these reactions are slow in aqueous solu-
agglutinate an entire panel of RBCs that had been tion at neutral pH. Penicilloyl derivatives arise
stored with penicillin as part of the preservative; RBCs optimally by direct acetylation of protein amino
from the same donors, when not exposed to penicillin, groups by the β-lactam carbonyl group at high pH
were not agglutinated by this serum. Watson and col- (e.g., pH 9–10), but several workers have shown that
leagues,36 in tests on 3000 unselected sera, found peni- penicilloyl determinants can form in vitro in neutral
cillin antibodies only in donors who had previously aqueous solutions.38
received penicillin. The immunologic behavior of penicillins (and proba-
Penicillin antibodies can be IgE, IgM, and IgG. IgE bly of cephalosporins) might be influenced greatly by
antibodies are rare; they are usually directed against the presence of impurities in the commercial products
minor determinants and are responsible for anaphy- and by changes that occur after the drug is prepared for
lactic reactions. IgM antibodies, usually directed injection. Polymers arising from intermolecular or
against the BPO determinants, are common. Using a intramolecular rearrangement of the parent antibiotic
sensitive passive hemagglutination technique, Levine can be present.38,39 In addition, proteins and polypep-
and coworkers37 detected penicillin antibodies in the tides derived from the fermentation process can be
sera of 98% of 57 random patients, and in 100% of 103 present in the commercial product; proteins can
patients who recently had received penicillin. IgM become heavily penicilloyed in the process.38 The
antibodies were detected in 98% of the former group amounts of polymer and protein vary considerably
and in 99% of the latter group; IgG antibodies were from product to product.39 It is interesting to note that
detected in 16% of the former group and in 38% of the Park and associates13 found that administration of ben-
latter group.37 Penicillin-induced cytopenias are zylpenicillin to rats at doses up to 1 g/kg did not lead
usually associated with IgG antibodies. Both IgG and to production of BPO antibodies, whereas a dose of
IgM penicillin antibodies are capable of activating benzylpenicillin one millionth of that amount, when
complement, and in vitro lysis of penicillin-treated conjugated to a foreign protein, was readily immuno-
RBCs can sometimes be observed with sera from genic. This observation suggests that contaminating
patients who have no evidence of hemolytic anemia protein in commercial penicillins could lead to
or a positive DAT. enhanced immunogenicity in some cases. Neftel40
Although penicillins bind well to proteins, the showed that intravenous administration of 200 million
mechanism (e.g., kinetics, catalytic factors involved, international units (IU) of penicillin G (one IU of peni-
role of cellular proteins vs. plasma proteins) involved cillin is the specific penicillin activity contained in 0.6 g
in the formation of the major immunogenic determi- of the crystalline sodium salt of penicillin G; 1 mg of
nants on proteins is not yet clear. penicillin G-Na = 1667 IU; 1 mg penicillin G-K = 1595
The basic structure of the penicillins is shown in IU) led to lymphocyte sensitization and IgG antibody
Figure 8-3. The structure consists of a thiazolidine ring production when the penicillin was dissolved and
connected to a β-lactam ring, to which a side chain (R) stored at 4°C for 4 to 24 hours before being injected, or
is attached. It is this side chain that differentiates the when the freshly prepared penicillin was given over a
various commercial penicillins. When animals, includ- period of 6 to 24 hours. Lymphocyte sensitization and
ing man, are immunized with benzyl penicillin, anti- antibody production were not seen when the penicillin
bodies are formed against a number of antigenic solution was freshly prepared and immediately given
Drug-Induced Immune Hemolytic Anemia 267

as a bolus. Neftel and colleagues41 later showed that far bodies react primarily with the hapten and either do
fewer reactions occurred in a group of 116 patients not react at all with the protein portion of the
given freshly dissolved penicillin. When intravenous complex or react only weakly. Hence, haptenic anti-
penicillin was given in the usual way to 193 patients, 7 gens are said to rarely, if ever, elicit “autoantibodies”
hemolytic anemias and 12 neutropenias were detected, capable of reacting with native autogenous protein.
in contrast to no such cytopenias after administration Shulman also presented experimental data indicating
of freshly prepared penicillin. Neftel and colleauges41 that a cell component is not part of the antigen.46-49 He
suggested that degradation and transformation pro- reported that concentrations of the drug on the order of
ducts formed in vitro, rather than the penicillin itself, 1 million times the concentration of cell sites for
were the causative agents. antibody attachment do not interfere with adsorption of
antibody. This is evidence that a cell-drug complex is
not the antigen; for if it were, free hapten in such great
The Immune Complex Hypothesis excess would be expected to inhibit the reaction
In 1952, Miescher and Miescher42 suggested that between antibody and the hapten-cell complex. More-
drug-immune complexes could explain apronalide over, by direct measurement of binding of the drug by
(Sedormid)-induced thrombocytopenia. They later cells, either in mixtures of cells and drug or by equili-
showed that rabbits immunized with foreign proteins brium dialysis, Shulman found that the association
had immune complexes attached to their platelets, between cells and drug was much too weak to account
leading to destruction of the platelets by macrophages. for the large amount of antibody that the same cells can
They suggested that a similar mechanism might be adsorb. On the other hand, drug antibodies could be
operating in drug-induced thrombocytopenia.43-45 shown to bind to drugs in the absence of cells.46-49
In a series of experiments using platelets and On the basis of these and other observations,
quinidine antibodies, Shulman46-49 provided support Shulman46-49 proposed that the immunologic idiosyn-
for this concept. Shulman reviewed the fact that the crasy that results in drug-induced cytolytic antibodies
sensitizing drugs that had been studied could be is the formation of high-affinity antibodies against a
removed from the cell surface by one quick saline stable complex of the drug with some soluble noncel-
wash—indicating that the cell-drug association is lular macromolecule (e.g., plasma proteins). He pro-
much weaker than the usual firm combination posed that the drug-induced antibodies reacted first
required for haptenic antigenicity. Further, he with the drug, and that subsequently, the antibody-
pointed out (as first mentioned by Landsteiner11) drug complexes were adsorbed nonspecifically by the
that when antibodies are formed as a result of an platelets (Fig. 8-4). The adsorption reaction, although
antigenic hapten-protein complex, they conceivably immunologically nonspecific in that it does not
could be directed against the hapten, the carrier involve an antigenic component of the cell, neverthe-
protein, or the entire complex; however, using less is selective for certain cell types and is a high-
platelet interactions, antibodies directed against the affinity reaction. Indeed, there is ample precedent for
entire complex had not been identified even though the involvement of cells in nonspecific immune
studies specifically designed in an attempt to adsorption phenomena, including experimental exam-
demonstrate them had been performed. Also, when ples of nonspecific cellular adsorption of antigen-
the antibodies are produced as a result of a union of antibody complexes.50-52 Later, the immune complex
hapten with autologous proteins, the resultant anti- hypothesis was extended to apply to RBCs.1,6,44

Complement

Complex formation

FIGURE 8-4. Proposed mechanism for immune complex-


mediated DIIHA. IgG drug antibodies are illustrated but the Drug
antibodies are often IgM. Drugs do not bond covalently to
RBCs, but rather complex with drug antibody. It is not known
whether complexes attach to the RBC by the drug portion or
the antibody portion of the complex, as illustrated at the top
and bottom, respectively, of the RBC shown at the right side
of the figure. The immune complexes activate complement.
CELL
Antibody
268 Immune Hemolytic Anemias

According to this so-called immune complex hypo- metabolite methimazole? Although the patient’s
thesis, intrinsic cellular components play no role in the serum reacted with RBCs in the presence of carbima-
development of the antibody. Therefore, this is not an zole, no reactions occurred if the patient’s serum was
instance of “partial autoimmunoreaction” as suggested allowed to incubate with the drug before RBCs were
by Ackroyd.30-34 Instead, the cell plays a purely passive added.55 One would not expect this if drug-antidrug
role and is, to use Dameshek’s53 vivid phrase, an “inno- immune complexes had been formed. In addition,
cent bystander.” Although “nonspecific” adsorption although the patient’s serum did not contain car-
was suggested by Shulman,46-49 one might suggest that bimazole, the drug could be detected on circulating
immune drug complexes could attach to the Fc recep- RBCs from the patient. Salama and colleagues55
tor on platelets or to the CR1 receptor on RBCs. interpreted these and other findings to indicate that
Miescher’s and Shulman’s “immune complex” a significant amount of carbimazole bound rapidly to
theory reigned supreme for 15–20 years, but then the RBCs in such a fashion that the molecular sites at
theory came to be questioned. In 1975, Ackroyd33 which hydrolysis takes place in the plasma were pro-
objected to the immune complex concept because the tected on the RBC membrane. This binding then
clinical effects observed in animals with immune com- might maintain the antigenic sites needed for
plexes on their platelets bears no resemblance to the immunogenicity and reactivity. In this case study, the
drug hypersensitivity syndrome observed in man. authors seem to have ruled out the possibility that
Ackroyd33 referred to work by Cronin,54 who immu- the drug or its metabolite combined with soluble
nized rabbits with apronalide (Apronal/Sedormid) macromolecules (carriers) in the plasma to induce an
conjugated to protein. He achieved this by substitut- immune response. They suggested that the drug
ing one of the amino groups of the apronalide mole- combined with the RBC membrane; antibodies were
cule with an amino-phenyl group. He then coupled induced by the drug-RBC complex, and these anti-
the amino-phenyl substituted compound to protein bodies reacted with the drug-RBC membrane
by diazotization. Rabbits immunized with this protein complex, activating complement and inducing in
conjugate developed precipitating antibodies to it. vivo hemolysis. It is also of interest that RBC autoan-
Precipitate formation was inhibited by unmodified tibodies, reacting against all reagent RBCs without
apronalide, showing that the immunologic reactivity the presence of the drug, were also eluted from the
of the apronalide molecule had not been altered by the patient’s RBCs, suggesting that some of the antibody
chemical manipulations employed. The rabbit anti- activity was directed solely against the RBCs. These
body had no effect on rabbit or human platelets in findings and hypotheses correlate closely with
vitro, however, even in the presence of apronal. Ackroyd’s work on platelets.30-34
Moreover, when the aminophenylated apronalide More findings that add to the controversy are the
compound was diazotized to human albumin, no following:
reaction could be demonstrated between this antigen
and the antibody in the serum of an apronalide- 1. Some drug-dependent antibodies, such as quini-
sensitive patient. These experiments, therefore, pro- dine antibodies, have been shown to bind to
vided no support for the hypothesis that the antibody platelets by their Fab domain.
in this type of drug hypersensitivity was stimulated 2. Many drug-induced antibodies appear to show
by a stable union of the drug with a soluble macro- specificity for certain cell lines or specific epitopes
molecule or that adsorption of drug-immune com- on platelets or RBCs.
plexes to platelets caused their lysis. 3. Many patients have now been described who
Salama and coworkers55 described a patient who seem to have antibodies with in vitro characteris-
developed acute intravascular hemolysis while tics of more than one of the previously described
receiving carbimazole (1-carbethoxy-3-methyl-2- mechanisms.
thiomidazole), a drug that was used for treating
hyperthyroidism. The drug rapidly hydrolyzes to DATA THAT DO NOT SUPPORT THE
methimazole, either in plasma or at an alkaline pH, IMMUNE COMPLEX HYPOTHESIS
and only the biologically active metabolite methima-
zole is detectable in plasma. The patient had IgG and Quinine/Quinidine-Dependent Antibodies Appear
C3 on her RBCs, and IgG autoantibody was elutable to Bind to Platelets by Their Fab Domains. Christie
from her RBCs. The serum contained carbimazole- and coworkers56 showed that platelets coated with
dependent antibodies reacting with untreated RBCs quinine- or quinidine-induced antibodies form
when carbimazole was added and also reacting with rosettes around protein A-Sepharose beads, and that
carbimazole-treated RBCs. The antibody did not normal platelets form rosettes around protein A-
react with the metabolite methimazole. This was Sepharose beads coated with these antibodies. These
notable, as carbimazole cannot be detected in plasma reactions occurred only in the presence of the sensitiz-
following oral administration due to its rapid ing drug. Platelets also formed rosettes about protein
hydrolysis to methimazole. This observation poses A-Sepharose beads coated with an anti-PlA1, but drug
the following question: How did the patient form was not required. Formation of rosettes between
antibodies to carbimazole but not to the circulating antibody-coated platelets and protein A-Sepharose
Drug-Induced Immune Hemolytic Anemia 269

was inhibited by F(ab’)2 fragments of goat antibody technique. 125I-F(ab′)2 fragments, prepared by pepsin
specific for the Fc portion of human IgG, whereas digestion of DEAE-chromatographed IgG and chlo-
rosette formation between antibody-coated protein ramine T labeling, bound to RBCs in drug-dependent
A-Sepharose and platelets was inhibited by F(ab’)2 fashion the same as drug-dependent IgG. Proof that
fragments directed against the F(ab’)2 portion of the the bound material was drug-dependent F(ab′)2 and
IgG molecule. Because binding of IgG to protein A is not a possible contaminant was obtained by eluting
known to occur via the Fc region, these findings sug- bound drug-dependent 125I-labelled protein from RBCs
gested that binding of drug-induced antibodies to and performing SDS-PAGE followed by autoradiogra-
platelets involve the Fab domains of the IgG molecule. phy. More than 90% of the eluted protein had a molec-
Using a different approach, Smith and associates ular weight of 100,000. The apparent association
(Shulman’s group)57 came to similar conclusions. The constant of the drug-dependent 125I F(ab′)2 binding was
antibody domain controlling reactions between approximately 2 × 108-1M and equal to that of intact IgG
platelet membranes and drug-dependent antibodies from which it was made. These results suggested a
from patients with thrombocytopenia induced by cin- similar Fab-dependent mechanism for RBCs as for
chona alkaloids were studied using F(ab’)2, Fab, and platelets.56-58
Fc fragments made from purified drug-dependent The foregoing data suggest that the drug antibody is
antibody. By direct-binding radioimmunoassay (RIA) reacting with an epitope on the cell membrane (e.g., a
measurements, 20,000–50,000 antibody molecules neo-antigen), not that immune complexes are attaching
bound per platelet equivalent of purified platelet “nonspecifically” to the membrane. Nevertheless, in
membranes at apparent saturation with three differ- 1993 Shulman and coworkers59 presented data against
ent antibodies. F(ab’)2 and Fab fragments bound to the concept of a neo-antigen. Their data suggested that
platelet membranes only in the presence of the drug; drug was the major target for antibody. They found, by
Fc fragments did not bind to platelet membranes. The equilibrium dialysis, that in reactions between purified
ability of drug-dependent IgG fragments to compete quinine or quinidine antibodies, platelet membranes,
with intact IgG was measured quantitatively by RIA and the drug, the amount of the drug in the complex is
and by complement fixation. F(ab’)2 and Fab com- proportional to the amount of antibody in the complex
peted with intact IgG at an 8:1 and greater than 50:1 and varies with drug concentration. At 10–8 M drug, the
molar ratio, respectively, by RIA, and at a 1.6–3:1 and ratio of drug to antibody is ~1:1 and affinity between
44–75:1 ratio, respectively, by complement fixation components of the complex is approximately 105 M–1,
assays. Fc did not compete with IgG in either assay. whereas the ratio approaches 2:1 and affinity approxi-
Smith and associates57 concluded that the Fab domain mately 108 M–1 as the drug is increased to 10–5 M. The
supported attachment of drug-dependent antibody to difference in affinity between reactants in the complex
the platelet membrane. at low and high drug concentrations is similar to the dif-
Jordan and colleagues (Shulman’s group)58 pre- ference in affinity between binding of drug-dependent
sented evidence that a tolmetin-dependent antibody Fab and F(ab’)2 at optimal drug concentrations. This
associated with severe intravascular hemolysis bound difference suggests that affinity and stoichiometry of
to RBCs by its Fab domain. Serum from a patient with complexes is controlled by antibody-bound drug and is
acute intravascular hemolysis was found to contain a consonant with the drug being bound avidly only in
drug-dependent IgM antibody that caused comple- complexes. Moreover, the glycoprotein (GP) sites reac-
ment fixation and agglutination, and a drug-dependent tive with drug-dependent antibodies do not appear to
IgG antibody that could be detected only by the IAT. be drug-specific-F(ab’)2 from quinine- or quinidine-
Jordan and colleagues58 found that the receptor for the dependent antibodies, which immunoprecipitate the
drug-dependent reaction was present on all RBCs of a same GPs (Ib/IX and/or IIb/IIIa), competitively
standard commercial panel and on Oh, Rhnull, Ko, inhibits fixation reactions of antibodies having the
Jk(a–b–), and i adult cells. Reactions of RBCs from cord opposite drug specificity. If neo-antigens were
blood were quantitatively the same as adult RBCs. involved, this would require the unlikely circumstance
Thus, none of these blood group antigens was the of drug isomers inducing separate neo-antigens at the
receptor for the tolmetin antibody. Solubilized RBC same GP site, at least two for each antibody specificity,
membranes were transferred by Western blot and less than 120 Ǻ apart. The researchers felt that their
incubated with drug-dependent antibody, followed by findings were consistent with the concept of the drug
a second incubation with either 125I-protein A or 125I- acting as a classical hapten. An initial low-affinity anti-
anti-IgG, all in the presence of the drug. The drug- body-drug reaction (Ka ≤ 105 M–1) might result in com-
dependent antibody receptor could not be identified plementarity between this binary complex and a GP
this way. But when 125I-labeled RBC membranes site, forming a ternary complex that could undergo
reacted with drug-dependent antibody were solubi- conformational change for increased stability.
lized, precipitated by staphylococcus protein A, eluted, Specificity for Certain Cell Lines or Specific Epi-
reduced, and electrophoresed in SDS-PAGE, they pro- topes on Platelets and RBCs. Drug-dependent anti-
duced an autoradiographic line primarily at the level of bodies appear to be highly specific for certain cell lines.
band 3. An antibody specific for band 3 produced the For instance, many drugs (e.g., quinidine, phenacetin)
same pattern as drug-dependent antibody by this have been documented to cause thrombocytopenia
270 Immune Hemolytic Anemias

and hemolytic anemia, but usually, only one cell line reacted with the patient’s serum only when D-positive,
is affected in a single patient. When two cell lines are M-positive RBCs were used to treat with streptomycin.
affected in one patient, the antibodies, reacting with It was said that streptomycin-treated D-negative, M-
each cell type, can usually be separated and prove to negative RBCs did not react, but no details were given
be from different antibody populations.48,60 One on how many different samples of various phenotypes
would not expect this if immune complexes were were tested. In 1981, Duran-Suarez and colleagues71
attaching “nonspecifically” to cells, but it might be described five examples of drug-dependent antibodies
explained if immune complexes were attaching pre- that would react with RBCs in the presence of the drug,
ferentially to the Fc receptor of platelets or to the CR1 but only when the RBCs were rich in I antigen (icord and
receptor of RBCs. iadult RBCs were nonreactive). The drugs involved were
Quinidine and quinine-dependent antibodies, reac- rifampicin, nitrofurantoin, and an antihistamine (dex-
tive with platelets, appear to react with specific chlorphenyramine maleate). Habibi and coworkers72
epitopes on platelet membrane glycoprotein (GP) and Salama and Mueller-Eckhardt73 also have found
Ib/IX and/or GPIIb/IIIa. In 1978, Kunicki and col- drug-dependent antibodies that reacted with adult
leagues61 showed that quinine/quinidine-dependent RBCs in the presence of the drug (thiopental anesthetic
antibodies failed to react with platelets from patients and nomifensine) and did not react with cord RBCs.
with Bernard-Soulier syndrome. This was confirmed Sandvei and associates74 described a drug-dependent
by others.60,62 Platelets from patients with Bernard- antibody that did not react with cord RBCs but reacted
Soulier syndrome are known to be deficient in the with iadult RBCs as strongly as Iadult RBCs (the authors
GP1b-IX complex, GPV, and at least one other protein. did not suggest it, but there is a possibility that the
Several investigators have produced evidence that specificity was anti-IT). In 1991, Pereira and colleagues75
quinine/quinidine-dependent antibodies react with described a single case of DIIHA and renal failure due
epitopes on the GP1b/IX complex.60,63-65 Christie and to rifampicin-dependent antibodies with anti-I
coworkers66 showed that some antibodies react with specificity. The antibodies reacted equally well with I-
GPIIb/IIIa. Visentin and associates67 studied sera positive RBCs of common phenotype or rare pheno-
from 13 patients with quinidine or quinine-induced types (Rhnull, pp, U–, Ko), but cord RBCs did not react.
thrombocytopenia. They found that 10 of 13 sera con- Habibi and Bretogne76 tested 19 antibodies, induced by
tained IgG antibodies specific for both GPIb/IX and 11 different drugs, against RBCs lacking high-incidence
GPIIb/IIIa, two reacted with GPIb/IX alone, and one antigens. Five antibodies did not react with D-negative
reacted with GPIIb/IIIa alone. In all cases, the drug RBCs. Two antibodies did not react with P-negative
was required for binding of IgG to target GPs. RBCs; five antibodies did not react with Lu(a–b–) RBCs.
Quinidine and quinine consist of linked quinoline and One antibody did not react with Ko RBCs. Only six anti-
quinuclidine ring structures and are lipophilic mole- bodies (32%) reacted equally well with all phenotypes.
cules. Visentin and associates67 suggested that as such Salama and Mueller-Eckhardt73 tested 30 nomifensine-
molecules are known to accumulate at amphophilic sur- induced antibodies against RBCs of common and rare
faces, they might concentrate preferentially in hydro- phenotypes in the presence of nomifensine. They found
phobic pockets within the chymotryptic-resistant that 14 of the 30 antibodies (47%) reacted with all RBCs
protein of the GPIIIa molecule, where they might tested. Nine antibodies (30%) were similar to drug-
induce structural changes (neo-antigens) that are independent autoantibodies, being nonreactive or
immunogenic in certain individuals. The researchers reacting more weakly with Rhnull RBCs. Three antibo-
suggested that antibody bound to such determinants dies (10%) did not react with cord RBCs and could be
might stabilize the drug-GP complex so that the drug inhibited by soluble I substance. The specificities of the
is not readily dissociated from the molecule by remaining four antibodies, although nonreactive with
washing. The finding of Christie and Aster68 that some RBCs, could not be identified. In 1984, Sosler and
tritiated quinidine is stabilized in the platelet mem- coworkers77 described a case of chlorpropamide-
brane by the binding of quinidine-dependent anti- induced IHA associated with Jka specificity. The
body is consistent with this possibility. patient’s serum contained drug-dependent anti-Jka that
It is interesting to note that Claas and coworkers69 showed enhanced reactivity in the presence of chlor-
found that none of ten drug-dependent antibodies propamide. Eventually, the anti-Jka was detectable only
associated with thrombocytopenia reacted with all in the presence of chlorpropamide (Fig. 8-5).
platelets in a panel from donors of different HLA In addition to some DIIHA-associated drug-
types. No HLA specificity could be determined, but induced antibodies showing blood group specificity,
the authors believed that genetic factors played a role. there are several examples of antibodies to chemicals
A number of drug-dependent antibodies have been that have shown blood group specificity. Beck78 and
reported to react with RBCs with blood group Dube and colleagues79 described caprylate-dependent
specificity. In 1977, Letona and associates70 reported a autoagglutinins of anti-c and -e specificity. Reviron
case of IHA induced by streptomycin. The patient’s and associates80 described an anti-I autoagglutinin
serum reacted with streptomycin-treated RBCs and that was enhanced in the presence of sodium azide. In
reacted with untreated RBCs when streptomycin was 1982, Halima and colleagues81 described an anti-Jka
added to the serum. The streptomycin-treated RBCs that at first appeared to be a low ionic strength saline
Drug-Induced Immune Hemolytic Anemia 271

128 believe that this phenomenon is due sometimes to the


64 presence of preformed drug-immune complexes in
Titer of antibody 32 the patient’s serum and that it can easily be differenti-
ated from true autoantibody. Once the drug is
16
stopped, the immune complexes are cleared rapidly
8 Chlorpropamide added from the plasma by the reticuloendothelial system;
4 No chlorpropamide added thus, the in vitro reactions caused by the drug com-
2 plexes are usually not seen with serum obtained
24 hours after drug cessation. In contrast, true autoan-
0 1 2 3 4 5 6 7 74 tibodies would still be present.
Weeks Florendo and colleagues89 described a patient
whose serum contained an RBC autoantibody that
FIGURE 8-5. The AGT scores of reactivity with Jk(a+) RBCs when chlor- was clearly separable from an antibody that reacted
propamide is added as compared with AGT scores without chlor-
propamide. (From Sosler SD, Behzad O, Garratty G, et al: Acute only with streptomycin-treated RBCs. Habibi and
hemolytic anemia associated with a chlorpropamide-induced auto coworkers90 described a patient with acute intravas-
anti-Jka.Transfusion 1984;24:206–209.) cular hemolysis and renal failure due to teniposide, a
semisynthetic derivative of podophyllotoxin used for
cancer chemotherapy. This patient’s serum reacted in
(LISS) solution-dependent antibody, but which was vitro with RBCs in the presence of teniposide to a titer
later determined both to react independently of ionic of 2024 but contained a weaker (titer of 8) RBC
strength and to be dependent on a preservative autoantibody that reacted without the presence of the
(paraben) added to the LISS. Paraben is a methyl ester drug. The autoantibody in this case was found to
of hydroxybenzoic acid. The anti-Jka would react with persist for 5 months, thus it could not have been
Jk(a+) RBCs only when methyl esters of hydroxyben- caused by the mimicking effect of immune complexes
zoic acid were added to the patient’s serum. The discussed previously. Bird and associates91 described
Jk(a+) patient had no signs of hemolytic anemia and two patients who developed positive DATs while on
was transfused with Jk(a+) RBCs with no ill effects.81 azapropazone. Both patients’ sera contained two dis-
Judd and coworkers82 reported three more examples tinct antibodies. One was an IgG autoantibody and
of similar nonpathogenic autoantibodies. It is difficult the other an IgM drug-dependent antibody.
to explain the foregoing blood group antigen drug- Habibi and colleagues72 and Salama and Mueller-
chemical antibody interactions on the basis of im- Eckhardt92,93 presented convincing data that more than
mune complexes attaching to cells. one mechanism can be operative in a single patient.
Serologic Findings Suggesting Different Mecha- Habibi and colleagues72 described four patients with
nisms Are Occurring in the Same Patient. Until rela- drug-induced acute hemolytic anemia and renal failure.
tively recently, most reports of individual patients The patients were shown to have drug-independent
with drug-induced IHA described antibodies showing antibodies to glafenine, latamoxef, and teniposide
characteristics associated with either autoantibodies together with drug-dependent RBC autoantibodies.
(e.g., methyldopa), the drug adsorption mechanism Salama and Mueller-Eckhardt92 studied 31 patients who
(e.g., penicillins), or the so-called immune complex developed IHA due to nomifensine. The majority of
mechanism (e.g., quinidine). Many reports now these patients’ sera (23 cases) contained IgG and/or IgA
suggest that antibodies showing characteristics of antibodies reacting to a highly variable extent with
more than one mechanism occur in individual RBCs, only in the presence of the drug and/or its
patients. Muirhead and associates83 described a case metabolites. Sera of six patients contained only IgG RBC
of acute hemolytic anemia associated with exposure autoantibodies, which reacted in the absence of the
to an insecticide containing chlorinated hydrocarbons. drug. Three patients had drug-dependent antibodies
The patient’s serum contained an RBC drug- and RBC autoantibodies in their serum. The patients
independent autoagglutinin and an antibody that with only autoantibodies, reacting by the “immune
caused in vitro hemolysis of RBCs sensitized with the complex” mechanism, experienced a less severe clinical
insecticide. In 1969, Hart and Mesara84 described a course than those with drug-dependent antibodies.
case of hemolytic anemia that appeared to be due to a Salama and Mueller-Eckhardt93 also described six
phenacetin-dependent antibody, but a drug-induced patients who had severe hemolytic anemia due to cian-
autoantibody was also thought to be present. It is not idanol, a flavoid used for treating hepatitis. Four
clear whether the antibodies, reacting with the patients developed drug-dependent IgM and/or IgG
patient’s own RBCs in these two reports, represented antibodies. One patient developed only IgG RBC
a true autoantibody. We (and others) have found that autoantibodies, and another patient developed a com-
sera from patients with drug-dependent antibodies, bination of IgG drug-dependent antibodies and IgG
reacting by the “immune complex” mechanism, some- autoantibodies. All drug-dependent antibodies reacted,
times can react with RBCs in vitro without the pres- in vitro, with RBCs in the presence of the drug (i.e.,
ence of added drug, thus suggesting the presence of typical of immune complex mechanism), but unex-
an additional drug-independent autoantibody.85-88 We pectedly, they also reacted with drug-coated RBCs (i.e.,
272 Immune Hemolytic Anemias

typical of the drug-adsorption mechanism). When the the drug to be present to demonstrate in vitro activity
drug was added to the patient’s serum in vitro (i.e., of the antibody). The other mechanisms discussed
hapten inhibition test), it did not prevent the reactions previously involved antibodies to drugs, and the
with drug-coated RBCs. RBCs became involved indirectly (e.g., either because
Salama and colleagues94 described two patients they became coated with drug or because the drug
with acute IHA caused by diclofenac. Both patients antibodies activated complement that reacted with
had developed IgG drug-independent antibodies and “innocent bystander” RBCs); in contrast, we are now
drug-dependent antibodies. The serum of one patient addressing drugs that seem to affect the patient’s
reacted with RBCs only in the presence of urine (ex immune system, causing a true autoimmune disease.
vivo antigen/metabolite) from patients receiving Throughout this section, we will use the term drug-
diclofenac. The serum did not react with RBCs in the induced autoantibody to describe those antibodies that
presence of diclofenac or its known metabolites. react with normal RBCs without the addition of
Serum from the other patient reacted with RBCs in the the drug in vitro, and which have no demonstrable
presence of ex vivo antigen and in the presence of the specificity for drug-related antigens (i.e., drug-
drug itself and of its main metabolite. The sera of two independent antibodies).
patients with tolmetin-induced IHA were shown to One of the most remarkable reports in the field of
contain drug-dependent and drug-independent anti- immunohematology was that of Worlledge and col-
bodies.95,96 Another closely related drug (zomepirac) leagues in 1966.105 Prior to their observations, the anti-
also led to production of drug-dependent and drug- bodies in all reports of DIIHA had specificity for
independent antibodies in a patient described by drug-related antigens, usually demonstrable only
Schulenburg and colleagues.97 when the offending drug, RBCs, and antibody were
Recently, the second- and third-generation cephalo- incubated together in vitro. In startling contrast,
sporins have been associated with severe DIIHA (see Worlledge and colleagues105 demonstrated that inges-
the discussion later in this chapter). Many of these tion of methyldopa resulted in the development of
cases have been associated with drug-induced anti- IgG autoantibodies and sometimes immune hemoly-
bodies that have characteristics associated with sis, in which the resultant antibody had demonstrable
several mechanisms.98-104 specificity for the patient’s RBC antigens but not for
Table 8-3 lists drugs that have been reported to be drug-related antigens. Serologic and clinical features
associated with antibodies suggesting characteristics do not differ from those found in patients with idio-
of more than one mechanism. pathic autoimmune hemolytic anemia (AIHA), and
therefore, the resultant anemia was appropriately
described as a drug-induced AIHA. Methyldopa-
RBC Autoantibodies Induced by Drugs induced immune hemolysis remains the most exten-
Sometimes drugs appear to cause a patient to produce sively studied drug-induced AIHA. Methyldopa is
RBC autoantibodies without the presence of drug- not commonly used now, but methyldopa-induced
dependent antibodies. These autoantibodies can cause AIHA will be reviewed in detail because it is the pro-
IHA and/or positive DATs without HA. The autoanti- totype drug for drug-induced AIHA and has been
bodies are drug independent (i.e., they do not require studied so thoroughly.

TABLE 8-3. DRUGS ASSOCIATED WITH ANTIBODIES SHOWING CHARACTERISTICS OF MORE


THAN ONE MECHANISM

Drug Mechanism* Reference

streptomycin AA + DA 70, 89
azapropazone AA + DA 91
teniposide AA + IC 90, 118
glafenine AA + IC 90
latamoxef AA + IC 90
nomifensine AA + IC 92
diclofenac AA + IC 94, 258
carbimazole AA + IC 56
tolmetin AA + IC 95, 96
zomepirac AA + IC 97
cefotaxime DA + IC 86
ceftazidime DA + IC 101
cianidanol AA + IC and AA + DA + IC 93
chlorinated hydrocarbons AA + DA + IC 83
carboplatin AA + DA + IC 167
cefotetan AA + DA and DA + IC and AA + DA + IC 87, 99, 100, 102, 104

AA, autoantibody; DA, drug adsorption mechanism; IC, “immune complex” mechanism.
Drug-Induced Immune Hemolytic Anemia 273

Possible Mechanisms for Methyldopa- Also, binding of methyldopa to RBCs has not been
Induced RBC Autoantibody Production demonstrated consistently. Although Green and
coworkers109,110 reported that methyldopa does bind
Because methyldopa-induced AIHA represents a to human RBC proteins, LoBuglio and Jandl111 incub-
human model for an important autoimmune disorder, ated 2-14C-α-methyldopa with RBCs and concluded
Worlledge and colleagues105 stated in 1966, “It is not that they were unable to demonstrate binding of the
too much to hope that the solution to the mechanism labeled drug to the RBC membrane.
of antibody formation in patients on methyldopa will Finally, this theory does not explain the more
lead to a better understanding of the causation of general autoimmune phenomena that are often asso-
AIHA and of autoimmune diseases in general.” ciated with methyldopa therapy, including antinu-
Unfortunately, this optimistic outlook has not been clear antibody production, chronic active hepatitis,
realized in its entirety, although a number of hypothe- and a lupus-like syndrome.6,7,112-117
ses have been presented and significant investigative Nevertheless, the hypothesis of alteration of RBC
work has been performed. membrane antigens has its proponents, in part because
Several clinical and laboratory findings of of observations strengthening the suggestion that
methyldopa-induced AIHA are difficult to explain drugs might always bind to antigens on cell mem-
because they are inconsistent with the usual kinetics branes even though such binding might be loose and
of antibody production in response to immunogens. difficult to demonstrate. Pertinent data were provided
For instance, the DAT usually does not become posi- by Habibi,118 who described patients who simulta-
tive until the drug has been administered for 3 to neously developed drug-dependent antibodies and
6 months, hemolytic anemia rarely occurs until RBC autoantibodies both having specificity for the
18 weeks after initiation of treatment, and the delay same well-defined blood group antigen. Habibi118
in onset of antibody production is not shortened pointed out that the simultaneous production of RBC
when patients who previously gave positive DAT autoantibodies and drug-dependent antibodies mimics
reactions are restarted on the drug—indicating that the well-known hapten and carrier specificities com-
there is no anamnestic response on repeated admin- monly developed in experimental animals immunized
istration of the drug. by hapten-carrier conjugates. He speculated that
In addition, the codevelopment of other autoanti- because both antibodies shared the same membrane
bodies (see the discussion that follows) strongly receptor, the formation of conjugates between drugs
suggests a more generalized immune reaction than and the RBC membrane must be the initial step of most
simply an alteration of the RBC membrane by the drug. DIIHAs in humans. Mueller-Eckhardt and Salama119
also have reported on numerous patients who simulta-
ALTERED AUTOANTIGENS neously developed drug-dependent antibodies and
drug-induced autoantibodies, some of which had the
One hypothesis suggested that the drug alters normal same specificity. They interpreted these findings to
RBC antigens in such a way that they are no longer rec- indicate that the drug most likely reacted with cell
ognized as self. Weigle106 demonstrated that tolerance membranes and that two types of antibodies might be
to antigens can be broken by injecting animals with self- produced.119 Drug-dependent antibodies could arise as
antigens that have been altered minimally. For example, a result of the production of a drug-dependent neo-
he produced thyroiditis and antibodies to native antigen that is composed of elements of both drug and
thyroglobulin by injecting rabbits with autologous cell membrane; drug-independent antibodies, which
thyroglobulin that had been picrylated or coupled can arise simultaneously in a given patient, could be
with diazonium derivatives of sulfanilic acid.106,107 elicited by a subtle alteration of antigens as described
Worlledge and coworkers,105 in their original report in by Weigle106,107 in his classic experiments. The binding
1966, postulated that the drug or one of its metabolites sites for these antibodies could be sufficiently similar to
might combine with the RBC membrane and alter Rh or could be composed of enough unaltered structures
antigens in such a way as to lead to the development of of normal blood cell membranes to support drug-
autoantibodies by the patient’s normal immune system. independent binding to both the patient’s cells and
The delay in development of a positive DAT could be normal cells.103,120 Such antibodies have the serologic
explained by the drug (or one of its metabolites) being characteristics of RBC autoantibodies, but we believe
incorporated into the RBCs at the normoblast or reticu- that they are formed as a result of mechanisms that are
locyte stage. Such RBCs would not necessarily have a different from those of autoantibodies induced by
reduced life span and would not reach the antibody- drugs (e.g., methyldopa or other drugs mentioned in
producing mechanism until at least 120 days had this section), which appear without the presence of
elapsed, thus explaining the delay in development of a drug-dependent antibodies.
positive DAT. As Worlledge108 pointed out, however,
this theory cannot explain the sometimes rapid reversal EFFECTS ON THE CELLULAR IMMUNE SYSTEM
in the DAT after the drug is stopped, because it implies
that there should be no significant change in the Dameshek121 suggested that methyldopa could have a
strength of the test for at least 3 months. direct effect on the cellular immune system. He
274 Immune Hemolytic Anemias

suggested that methyldopa produces an aberration in 3. It inhibits T-cell DNA methylation, inducing T-cell
lymphocyte proliferation leading to the emergence of autoreactivity.130
autoantibody-producing cells. This proposed mecha- 4. It interferes with immunoregulation.131-136
nism is consistent with the prolonged period of time
necessary to induce immunohematologic abnormali- The reported effects on cellular immune function
ties in patients, even on repeated challenge with the conflict with one another, and the described effects
drug.108,122 Although a significant body of experimen- were small, requiring relatively high concentrations of
tal work has been developed with regard to this procainamide. Ochi and colleagues137 showed that
proposed mechanism of autoantibody production, the procainamide impaired generation of suppressor T-
studies have led to inconclusive results. cell activity but that it exerted no enhancing effect on
Kirtland and associates123 measured cyclic AMP pro- T-helper cell, B-cell, or macrophage activities. Some
duced in vitro by lymphocytes from healthy donors members of this same group later published findings
after adding methyldopa, and by lymphocytes from that conflicted somewhat with their previous conclu-
patients who were receiving methyldopa. Significantly sions.136,137 They found that patients on long-term
higher lymphocyte cyclic AMP concentrations were procainamide therapy had normal numbers and ratios
generated by both sets of lymphocytes compared with of helper and suppressor T cells and normal mitogen-
lymphocytes from healthy donors without methyldopa induced suppressor cell activity. A significant reduc-
present. To measure the effect of methyldopa on sup- tion in mitogen-induced IgG secreting cells was
pressor cells, Kirtland and associates123 used an assay of attributed to a decrease in both T-helper and B-cell
suppressor cell activity based on the finding that prein- activity in 50% of the patients and to only a B-cell
cubation of lymphocytes before mitogen stimulation activity decrease in 25% of the patients. The authors
in culture leads to less IgG being generated by suggested that the defects in B- and T-cell function
B cells. This is purportedly due to enhancement of sup- could result from the ability of procainamide to inhibit
pressor T-cell activity after the in vitro preincubation membrane depolarization by a mechanism similar to
phase.123 Kirtland and associates123 confirmed the that observed in the cardiac conduction system.138
results of Lipsky and colleagues124 that less IgG was Procainamide could nonspecifically suppress lym-
produced after preincubation of lymphocytes. They phocyte function through the action of the hydropho-
showed that if methyldopa was added during the bic parts of the molecule, analogous to the anesthetic
preincubation phase, the inhibition of IgG generation effect of its analogue, procaine.138
during mitogen-stimulated culture was negated. The Miller and Salem134 found normal suppressor cell
amount of IgG generated in vitro after adding methyl- function in 14 patients receiving procainamide. Total
dopa to the preincubation phase was significantly in vitro IgG generation from their mitogen-stimulated
greater than the IgG generated without a preincubation lymphocytes was significantly increased, however,
phase. Similar differences were observed when lym- compared with healthy controls and patients with sys-
phocytes from patients receiving methyldopa were temic lupus erythematosus (SLE). Separated T cells
compared with lymphocytes from healthy donors. from patients taking procainamide did not affect in
These results were interpreted to show that methyldopa vitro suppressor function of T cells from healthy
interfered with the normal function of suppressor T individuals. The authors postulated that procai-
cells to moderate IgG autoantibody production by namide induced autoantibodies by enhancing helper
B cells. Using similar techniques to those used by T-cell function rather than impairing suppression.
Kirtland and associates,123 Garratty and colleagues125 DeBoccardo and associates135 found that procain-
confirmed the effect of preincubation on in vitro IgG amide inhibited in vitro IgG secretion and generation
generation by B cells reported by Lipsky and of IgG plaque-forming cells. The drug inhibited dif-
coworkers,124 but they could not confirm the findings ferentiation of B cells to plasma cells rather than pro-
of Kirtland and associates123 that methyldopa had duction and secretion of IgG. The authors postulated
any effect on suppressor functions, as defined by the that procainamide inhibits mitogen-induced B-cell
in vitro production of IgG after a preincubation maturation to plasma cells by inhibiting production of
phase. Garratty and colleagues125 agreed with the cytokines by helper cells. These results contrasted
findings of Kirtland and associates123 that methyl- with those reported by Ochi and colleagues,137 but it
dopa depressed the proliferative response of should be pointed out that DeBoccardo and asso-
mononuclear cells to mitogen stimulation. ciates135 used much higher concentrations of procain-
Procainamide also induces autoantibodies, includ- amide than those used by Ochi and colleagues,137
ing RBC autoantibodies.126,127 The following hypo- which were within the usual therapeutic plasma
theses have been proposed for the induction of range. Bluestein and coworkers132 found a biphasic
autoantibody production by procainamide: response when they studied the effect of different
concentrations of procainamide on mitogen-induced
1. It interacts with nucleoprotein to form a neo- lymphocyte proliferation. Marked suppression was
antigen.128,129 observed at a high concentration of the drug, but at
2. It acts as an adjuvant for polyclonal activation of lower doses of the drug, enhanced proliferation was
autoantibody-producing clones. observed. The lower concentrations of procainamide
Drug-Induced Immune Hemolytic Anemia 275

were nearer those found in plasma following therapy. drug-independent binding to both the patient’s cells
Garratty and colleagues125 found that therapeutic and normal cells (drug-independent neo-antigen). Such
levels (30 μg/mL) of procainamide did not signifi- antibodies behave like autoantibodies and cannot be
cantly affect PHA-induced lymphocyte proliferation. distinguished from “true” autoantibodies (i.e., those
This result did not agree with the results of Bluestein associated with warm AIHA). Salama and colleagues94
and coworkers133 but agreed with those of Ochi and believe that only one hypothesis is necessary to explain
colleagues,137 who reported that procainamide con- all the phenomena we observe. They not only criticized
centrations ranging from 10 μg/mL–40 μg/mL had no the immune complex hypothesis but also criticized the
significant effect on PWM-induced proliferation as concept of the drug-adsorption mechanism and the
measured by tritiated thymidine incorporation. Using theory that some drugs (e.g., methyldopa and pro-
the assay described by Lipsky and associates124 and cainamide) cause autoantibody production by affecting
Kirtland and coworkers,123 Garratty and colleagues125 the immune system directly.
found that procainamide did not affect suppressor cell It is tempting to draw cartoons as Habibi118 and
activity. These results agree with those of Miller Garratty103,120 did to illustrate how the immune
and Salem,134 DeBoccardo and colleagues,135 and Yu complex, drug adsorption, and autoantibody mecha-
and Ziff136 but do not agree with those of Ochi and nisms could be explained on the basis of classical hapten
coworkers.137 immunology (e.g., Fig. 8-6). These cartoons are based on
the findings that in the haptenic response, antibodies
can be made to the hapten (i.e., drug), the carrier (e.g.,
Newer Concepts of the Immune RBC membrane components), and/or a population of
Response to Drugs Associated antibodies that could react with part hapten, part mem-
with Cytopenia brane. One could argue that a patient might make one
or several populations of antibodies showing the differ-
To explain why the sera of four patients with drug- ent specificities. Thus, an antibody to the drug alone
induced IHA showed the characteristics associated with would react with drug-coated RBCs and be inhibited by
the “immune complex” and autoantibody mechanisms, the drug; this would be typical of penicillin antibodies
Habibi118 suggested a single mechanism. He suggested or the “drug adsorption” mechanism. Antibodies to
that after ingestion of a particular drug, the formation of membrane components would appear only as autoanti-
both autoantibodies and drug-dependent antibodies bodies. Antibodies directed to epitopes that are part
could be explained by the well-known hapten and drug/part membrane would require drug and RBCs to
carrier specificities commonly developed in animals be present together before a reaction is observed.
immunized with hapten-carrier conjugates. He sug- Because most of these drugs do not combine with RBCs
gested that formation of drug-cell conjugates must be efficiently enough to withstand in vitro washing, one
the initial step of most drug-induced cytopenias, and cannot prepare drug-coated RBCs, and the only way of
that the rare incidence of this disorder is probably demonstrating such antibodies is to mix the patient’s
because only few individuals are capable either of serum with drug and RBCs. This reaction is typical of
coupling drugs to their cells in vivo to form efficient the so-called immune complex mechanism, but if the
immunogens or of mounting an unusually strong foregoing concept is correct, the antibody is reacting
immune response to such conjugates. Salama and col- with a neo-antigen that is formed when the drug binds
leagues94 suggested a unifying concept that is basically loosely to cell membrane components, rather than drug
similar to that of Ackroyd30-34 and Habibi118 regarding immune complexes attaching to the cell. Extending the
the proposed immune response to drugs. They sug- suggestions of Habibi118 and Salama and Mueller-
gested that the immune process is always initiated by a Eckhardt,94 Garratty103,120 suggested that sometimes the
primary interaction of the drug and/or its metabolites epitope inducing the immune response might be par-
with constituents of blood cell membranes. This inter- tially drug and partially membrane protein, but prima-
action provides the composite antigenic structure, rily membrane protein (see Fig. 8-6). In this case, the
which provokes the production of two types of anti- drug might not be needed to demonstrate the antibody
bodies—drug-dependent and/or drug-independent. in vitro, and the antibody might appear to be a drug-
The specificity of drug-dependent antibodies is de- independent antibody (autoantibody).
termined by elements of both drug and cell membrane The unifying hypothesis is attractive because it can
(drug-dependent neo-antigen). These antibodies cannot be used to explain the Fab-dependent binding of the
bind sufficiently well to either drug or cell membrane drug antibody, the specificity that is sometimes
alone. If one part is removed (e.g., by dialysis in vitro, observed, and the serologic finding that suggests
by discontinuance of drug administration, or by subse- several mechanisms in one patient. When the antibody
quent excretion in vivo), the immune reaction subsides. is made against part drug/part membrane, the propor-
The authors suggested that drug-independent antibo- tions recognized as antigenic determinants will vary.
dies could be elicited by a subtle alteration of the mem- For instance, one population of antibody molecules
brane by the drug, but their binding sites are might be mainly antidrug, and another population
sufficiently similar to (or are composed of enough un- might be mainly antibody directed at membrane pro-
altered structures of) normal cell membranes to support teins (as drug plus membrane proteins) (see Fig. 8-6).
276 Immune Hemolytic Anemias

Antibody to drug relatively commonly, and these are not seen together
with antibodies showing characteristics of the
Antibody to immune complex or drug adsorption mechanism.
(mainly) membrane
components Drug Antibody to drug and We believe that the drug-independent antibodies
membrane components induced by drugs such as methyldopa, fludarabine,
and procainamide are formed by a mechanism differ-
ent from the mechanism illustrated in Figure 8-6.
These drugs always lead to the development of
autoantibodies and are never accompanied by drug-
independent antibodies. In contrast, other drug-
Red cell membrane independent antibodies (e.g., nomifensine and the
FIGURE 8-6. Proposed unifying hypothesis of drug-induced antibody newer cephalosporins) almost always occur together
reactions. The thicker, darker lines represent antigen-binding sites on with drug-dependent antibodies. This observation
the Fab region of the drug-induced antibody. Drugs (haptens) bind fits well with the concept (illustrated in Fig. 8-6) that
loosely (or firmly) to cell membranes, and antibodies can be made to several populations of antibodies are likely to be
(a) the drug (producing in vitro reactions typical of a drug adsorption
[penicillin-type] reaction); (b) membrane components, or mainly mem-
formed. Although it is possible for only one popula-
brane components (producing in vitro reactions typical of autoanti- tion to be formed in an individual patient, it is hard
body); or (c) part-drug, part-membrane components (producing an in to accept that methyldopa, fludarabine, and pro-
vitro reaction typical of the so-called immune complex mechanism). cainamide always induce only a single population
(From Garratty G: Target antigens for red-cell bound autoantibodies. In reacting primarily with membrane components.
Nance SJ (ed): Clinical and basic science aspects of immunohemato-
logy. Arlington, VA: American Association of Blood Banks, Although the findings of Garratty and colleagues125
1991:33–72.) suggest that methyldopa and procainamide do not
affect suppressor cell function as measured by
Kirtland and associates,123 we believe that these drugs
Garratty103,120 suggested that the membrane compo- do affect immune function in some way to allow pro-
nents composing the immunogen might contain liferation of true autoantibody.
specific, well-defined platelet or RBC antigens and Shulman and Reid139 have put forward a defense of
that the “drug” antibody, therefore, will appear to the immune complex concept and have incorporated
have blood-group specificity. Because the proportion some aspects of the unifying hypothesis. They illustrate
of membrane and drug antigenic determinants vary, their article with several cartoons (Figs. 8-7 and 8-8)
one would expect to see a spectrum of serologic activ- that combine elements of the original immune complex
ity among different patients. Thus, patients said to concept and the unifying hypothesis. They agree that
have antibodies showing characteristics of the drug antibodies (other than those related to penicillin)
immune complex, drug adsorption, and autoantibody are induced by antigens formed when the drug binds
mechanisms could have three different antibody pop- loosely to the cell in vivo. They point out, however, that
ulations, but the serologic characteristics could be most experiments of hapten immunology have
explained by a single mechanism. involved injecting animals with preconjugated hapten-
protein complexes. Because drugs other than penicillin
Problems with the Unifying Hypothesis do not form covalent bonds rapidly with proteins in
vivo, most drug antibodies are probably formed
The unifying hypothesis is based on classical hapten against metabolites of the original drug. These authors
immunology studies performed by injecting into suggest that penicillin antibodies could react with pre-
animals small MW substances that conjugated in vitro formed drug-cell complexes, but that antibodies to
to heterologous proteins (e.g., albumin). In a review in other drugs (e.g., quinidine/quinine) could react with
which they propose a variation on the original drugs bound loosely to the cell membrane or with a
immune complex hypothesis, Shulman and Reid139 drug that is not bound to cell membranes. In either
point out that when autologous or homologous case, one could say that a drug-antidrug immune
protein (rather than heterologous protein) is used as a complex is formed. To explain the Fab binding of drug
carrier, the complex is less immunogenic, and autoan- antibody to the cell, Shulman and Reid139 suggest that
tibodies rarely develop. Thus, the claims that the “uni- drug-immune complexes could attach to the cell by the
fying” hypothesis is supported by classical hapten weak attraction of the drug to the cell, or the antibody
immunology is not strictly true, considering that all might bind to drug bound loosely to the cell. If the drug
the situations we are discussing involve autologous antibody also recognizes some membrane antigens
protein carriers. As drug-induced IHA is so rare, it (e.g., neo-antigens), then the binding of the complex to
might be suggested that when we observe mixed sero- the membrane would be stronger. Finally, Shulman and
logic characteristics suggesting more than one mecha- Reid139 suggest that conformational changes in the Fab
nism, these could be rare examples that do not follow portion of the drug antibody might occur after drug
the usual hapten immunology and could be explained binding, and that such conformational changes might
by the unifying hypothesis. We find it hard to accept recognize membrane sites. Such interactions could
that the unifying hypothesis explains autoantibodies convert a low-affinity reaction (i.e., antibody reacting
induced by drugs such as methyldopa, fludarabine, with loosely bound drug) to a higher- (but still low-)
and procainamide. Such drugs induce autoantibodies affinity bimolecular association. If two of the drug
IHA-08(261-318) 11/18/03 4:43 PM Page 277

Drug-Induced Immune Hemolytic Anemia 277

TYPES OF SEQUENTIAL REACTIONS


POSSIBLY LEADING TO HIGH AFFINITY COMPLEXES

Bimolecular Low affinity High affinity


low affinity transitional conformational
reaction complex stabilization

Drug-
A antibody
complex

or

Drug partitioned
B by amphipathic
cell protein

Drug partition site


C similar to covalent
binding site

Conformational
D change at
drug partition site

FIGURE 8-7. Possible rearrangements of transitional reactions leading to high affinity complexes. Note absence of covalent bond between drug and
protein. (From Shulman NR, Reid DM: Mechanisms of drug-induced immunologically mediated cytopenias. Transfus Med Rev 1993;VII:215–229.)

Drug
Cell membrane
Fab

Ka < 5 × 104
Ka < 3 × 105

Low affinity
bimolecular
A associations
(Conformational
change in Fab)

Trimolecular
B association
(Diffusion
Ka ∼ 5 × 106 limitation)
Ka ~ 108

F(ab)2 or lgG
C

High affinity
stabilization
(Conformational
change in Fab + cell)
FIGURE 8-8. Possible changes in Fab and cell membrane protein leading to monovalent and divalent high-affinity complexes. (From Shulman NR, Reid
DM: Mechanisms of drug-induced immunologically mediated cytopenias. Transfus Med Rev 1993;VII:215–229.)
278 Immune Hemolytic Anemias

antibody’s Fab sites [F(ab)2] are involved in the interac- system in a similar way to RBCs coated with IgG RBC
tion, this process could convert a low-affinity monova- blood group auto- or alloantibodies (see Chapter 4).
lent reaction to a divalent high-affinity interaction (see Penicillin can be detected on the surface of the RBCs
Figs. 8-7 and 8-8). of most patients who are receiving high doses of the
drug intravenously.141-145 Using a rabbit penicillin
antibody, Levine and Redmond144 demonstrated
SUGGESTED MECHANISMS OF penicillin on the RBCs of 30% of the patients taking
DRUG-INDUCED HEMOLYTIC ANEMIA 1.2–2.4 million units per day and on the RBCs of all
AND/OR POSITIVE DAT patients taking 10 million units or more daily. This
coating is not by itself injurious, but some patients
develop a high-titer penicillin antibody that reacts
Possible pathogenic mechanisms are closely related with RBC-bound penicillin. Large doses of the drug
to the hypotheses discussed previously for the appear to be necessary to coat RBCs sufficiently to
immune response to drugs. The cartoons used (e.g., furnish enough antigenic sites for reaction with peni-
Figs. 8-4, 8-6, 8-7, 8-8, and 8-9) reflect on both the cillin antibody. Thus, the mechanism of cell sensi-
suggested epitopes that are thought to stimulate tization and resultant hemolysis differs from those
drug antibodies and their reactions with drug and discussed previously; it appears to result from the
RBCs in vivo and in vitro. coating of RBCs by penicillin during the administra-
tion of high doses of the drug, and from the reaction
Penicillin-Type Mechanism of penicillin antibodies with such drug-coated RBCs
(see Fig. 8-9). The quantity of penicillin antibody sen-
This mechanism is the best understood and the least sitizing the RBC is related to the concentration of peni-
controversial; it has also been called the “hapten mech- cillin antigenic determinants on the RBC membrane,
anism” and the “drug adsorption mechanism.”140 The the plasma concentration of penicillin antibody, and
term “hapten mechanism” implies that the hapten-like the avidity of the antibody.
binding of penicillin to RBCs is essential for the drug to When sensitive techniques are used, penicillin anti-
be immunogenic, as was postulated by Ackroyd.30-34 bodies can be detected in more than 90% of patients’
There is no evidence, however, that RBC binding by sera. Most sera in the study by Levine and collea-
penicillin is necessary for the drug to be immunogenic. gues37 contained IgM antibodies; the sera of approxi-
Instead, the immunogenicity of penicillin could relate mately 16% of random patients and 38% of patients
to its binding with plasma proteins, and hemolysis who had received penicillin recently contained IgG
could result from the reaction of penicillin antibody antibodies also. These antibodies were usually neu-
with penicillin that has been adsorbed to the RBC tralized by BPO hapten. The antibodies associated
membrane (this is why we termed it the “drug adsorp- with IHA due to penicillin are IgG, and these power-
tion” mechanism1,140). ful antibodies are not easily neutralized by BPO
Some drugs (e.g., penicillins and cephalosporins) hapten; inhibition is observed if dilutions of the high-
bond covalently to proteins (e.g., proteins on cell mem- titer penicillin antibody are used for the inhibition
branes) (see Table 8-2). When patients receive large tests. The high incidence of penicillin antibodies
enough doses (e.g., several million units of penicillin detected in random patients (and in healthy individu-
per day) of such a drug, the RBCs become coated with als) is probably due to the continual exposure to peni-
the drug, and any antibody the patient makes might cillin in our modern environment.
attach to the drug on the RBCs (see Fig. 8-9). When these Theoretically, antibodies to any of the drugs that
antibodies are IgG, the IgG/drug-coated RBCs could be bind firmly to RBCs (see Table 8-2) could cause a
removed by macrophages in the reticuloendothelial hemolytic anemia through a mechanism similar to

Antibody

CELL

FIGURE 8-9. Mechanism of penicillin-induced DIIHA and/or


positive DAT. Penicillin bonds to RBCs, and IgG penicillin anti-
body attaches to RBC-bound penicillin.

Drug
Drug-Induced Immune Hemolytic Anemia 279

that described for penicillin. In practice, most such


antibodies appear to cause hemolytic anemia through TABLE 8-4. CHARACTERISTIC FEATURES OF
other or additional mechanisms. It could be that the PENICILLIN-INDUCED HEMOLYTIC ANEMIA
classical penicillin mechanism is dependent on very
Hemolysis develops only in patients who are receiving large doses of
large amounts of drug being given. penicillin intravenously (at least 10 million units daily in an adult
It is possible for penicillin antibodies to activate for a week or longer).
complement, and on rare occasions this process could Hemolytic anemia is less acute in onset than that caused by the
be efficient enough to cause acute complement- immune complex mechanism, and it usually develops over a
mediated intravascular hemolysis.146-148 This probably period of 7 to 10 days. Hemolysis can be life-threatening,
however, if the etiology is not promptly diagnosed and if penicillin
occurs in addition to the IgG-mediated destruction of administration is continued.
RBCs by macrophages. The DAT is strongly positive due to RBC-bound IgG; about 45% of
Table 8-4 shows the characteristic features of patients could have RBC-bound complement detected as well.
penicillin-induced hemolytic anemia. Antibody eluted from the patient’s RBCs reacts with penicillin-treated
RBCs but not with untreated RBCs.
A high-titer IgG penicillin antibody is present in the serum. Using a
Nonpenicillin-Type Mechanism high-pH method to coat RBCs described later in this chapter, the
titer is usually 1000 or greater.
(“Immune Complex” Mechanism) Cessation of penicillin therapy is followed by complete recovery,
although hemolysis of decreasing severity and a “mixed-field”
Most of the drugs that have caused DIIHA do not cova- positive DAT can persist for several weeks.
lently bind to RBCs, and one cannot prepare drug- Other manifestations of penicillin allergy are not necessarily present.
coated RBCs in vitro to test for drug antibodies. As
discussed in the section on the immune response to
drugs, it is controversial how such drugs evoke an
immune response, and it is equally controversial how Regardless of which theoretic concept is correct, the
the drug antibodies interact in vivo to cause cytopenia laboratory and clinical findings associated with this
(e.g., hemolytic anemia). It is obvious that most of group of drugs are quite distinct and contrast with the
them, in contrast to penicillin-type antibodies, cause penicillin-type mechanism. Table 8-5 lists the charac-
cytopenia by complement-mediated reactions. The teristics of these drug-dependent antibodies.
hemolytic anemia is often acute, severe, and associated
with hemoglobinemia and hemoglobinuria; many are Drug-Induced Nonimmunologic
associated with renal failure. Early work on drug-
induced thrombocytopenia suggested that this group Adsorption of Protein onto RBCs
of drugs could sometimes evoke an antibody (often Some drugs appear to cause proteins to adsorb onto
IgM) that binds efficiently to drug-forming immune RBCs by a nonimmunologic mechanism, leading to a
complexes in vivo. These immune complexes would positive DAT. The first drug to be described as causing
attach to platelets nonspecifically (or perhaps to the Fc this effect was cephalothin.149-151 Other cephalo-
receptor on platelets) and activate complement. It was sporins might be capable of the same effect. Molthan
suggested that only a small amount of drug (or and coworkers149 reported that 81% of 31 patients
immune complex) would be needed to activate com- receiving cephalothin had a positive DAT; Gralnick
plement and that the immune complexes would be able and associates150 and Perkins and colleagues151
to move from cell to cell, activating complement, thus reported the same finding in 40% of 20 patients and
creating an efficient way of damaging many cells. This 38% of 143 patients, respectively. In an extensive
would be in contrast to the penicillin mechanism, study in 1971, Spath and coworkers152,153 found that
where large amounts of drug are needed to coat only 4% of 320 DATs performed on 97 patients receiv-
enough RBCs to react efficiently with the IgG penicillin ing cephalothin were positive. They suggested the
antibody. This mechanism has been called the immune existence of several explanations for the difference
complex mechanism (see Fig. 8-5) and also has been between their results and those of other researchers.
used to explain some types of DIIHA. The clinical and
serologic findings associated with these types of anti-
bodies certainly seem to fit with immune complex- TABLE 8-5. CHARACTERISTICS ASSOCIATED
mediated in vivo and in vitro reactions (Table 8-5). WITH DRUG-DEPENDENT ANTIBODIES OTHER
Nevertheless, as discussed previously, the immune THAN THE “PENICILLIN TYPE”
complex mechanism as originally described has been
criticized strongly, and it has been suggested that it Patient need take only a small amount of the drug.
should be replaced by a mechanism that involves an Acute complement-mediated hemolysis often occurs; 30%–50% of
interaction of the drug antibody against a neo-antigen. patients have associated renal failure.
The patient’s RBCs are often sensitized with complement only, but
This neo-antigen could be a new antigen produced by a RBC-bound IgG and/or IgM can be present.
chemical change in the cell membrane induced by brief Patient’s serum reacts with RBCs in the presence of the drug and/or
(i.e., noncovalent-binding) contact with the drug, or the its metabolite. Antibodies are often IgM, but IgG antibodies can
new antigen could be a combination of part drug part be present alone or together with IgM. Antibodies can cause
membrane protein. The latter explanation is the basis of hemolysis, agglutination, and/or sensitization of RBCs in the
presence of drug.
the unifying hypothesis discussed previously (see After drug is stopped, hematological remission is usually rapid.
Fig. 8-6).
280 Immune Hemolytic Anemias

Nonimmunologic adsorption of proteins is usually Although Spath and coworkers152 confirmed that
suspected when most normal plasmas/sera react (e.g., cephalothin can cause nonimmunologic adsorption of
by IAT) with drug-treated RBCs but eluates from the protein, three of four patients with a positive DAT
RBCs are nonreactive. After such findings, we usually (but no hemolytic anemia) had demonstrable IgG
confirm our suspicions by performing an antiglobulin anticephalothin on their RBCs. Thus, most of the posi-
test with an antihuman albumin that has been tive DATs appeared to be due to an immune mecha-
standardized for use with RBCs. The presence of RBC- nism, although additional nonimmunologic uptake of
bound albumin is strong evidence for nonimmuno- protein was not excluded. In a companion paper,
logic uptake of plasma proteins. IATs using antisera to Spath and coworkers153 studied optimal conditions
other proteins are also often positive, as demonstrated for detecting cephalothin and penicillin antibodies.
by Spath and coworkers.152 The studies confirmed earlier work indicating that
Patient selection can influence the results greatly. The penicillin binds optimally to RBCs at around pH 10. A
dosage and duration of therapy are likely to influence penicillin antibody reacted to a titer of 200, 400, and
the incidence of positive reactions. The studies of Spath 3200 with penicillin-coated RBCs prepared at a pH of
and coworkers152 were carried out in consecutive un- 7.3, 8.2, and 10, respectively. A cephalothin antibody
selected patients in a community hospital, and neither with a titer of 3200 showed no significant difference
the mean daily cephalothin dosage (6.3 g), the mean when RBCs were coated with cephalothin at pH 7.3,
duration of therapy (5.5 days), nor the incidence of renal 8.2, or 10.153
insufficiency was high. The distribution of BUN levels Molthan and colleagues149 and Gralnick and associ-
in their patients was as follows: 80% normal levels, 12% ates150 suggested that the cephalothin-induced DATs
in the range of 20–30 mg%, 4% between 30 mg% and were due to cephalothin-protein conjugates attaching
50 mg%, and 4% higher than 50 mg%. Three of the four to the RBCs and that antiglobulin sera reacted with
patients with positive DATs in their series had elevated RBC-bound protein. Spath and coworkers152 showed
BUN levels. A greater incidence of positive DATs might quite conclusively that the suggestions of these other
logically be anticipated in referral centers or infectious authors were not correct. Spath and coworkers152
disease units caring for more seriously ill patients. This showed that washed cephalothin-treated RBCs adsorb
might be especially true if a large percentage of patients a variety of proteins that react with antiglobulin sera
studied have renal disease, as in the reports of Molthan (i.e., preformed cephalothin complexes are not neces-
and colleagues,149 Gralnick and coworkers,150 and sary to obtain positive antiglobulin tests). In 1975, we
Perkins and associates.151 In addition, such patients suggested that cephalothin bonds (like penicillin) to
could have a positive DAT unrelated to cephalothin the RBC membrane, but that unlike penicillin,
therapy or because drug is not excreted efficiently, cephalothin changes the RBC membrane, allowing
leading to high levels in the plasma. Gralnick and proteins to attach to the membrane (Fig. 8-10).140 It
coworkers150 and Perkins and associates151 did not was not clear how the proteins attach to the modified
report results of pretreatment DATs and did not present membrane.
specific immunologic data indicating that the positive Petz and Branch14 and Branch and associates154
DATs were caused by cephalosporin administration. reported that some cephalosporins (cephalexin, cefa-
Thus, in any series of patients and particularly among zolin, cefamandole) did not cause nonimmunologic
patients with renal disease, control series indicating the binding of protein to RBCs. They suggested that the
incidence of positive DAT in a similar group, pretreat- nonimmunologic adsorption of protein might not
ment antiglobulin tests, or specific immunologic data in occur because the RBC membrane is changed by the
each patient (such as the elution of specific antibody cephalosporin, and that cephalothin might bind to
from the patient’s RBCs) are necessary before the posi- RBCs not through its β-lactam group but rather
tive DATs can be attributed to cephalosporin therapy. through acetoxymethylene and/or thiopene side
The nature of the antiglobulin sera (AGS) could be of chains.154 An alternative suggestion was that RBCs
significance. To ensure that these results might be might have a specific receptor site that remains active
readily duplicated by others, Spath and coworkers152 under mild acid conditions. If either of these events
performed the initial antiglobulin tests with a readily occurred, then perhaps the exposed β-lactam groups
available commercial polyclonal polyspecific AGS that on cephalothin would be free to react with plasma
had previously been evaluated and shown to possess proteins. This would explain why, under acid condi-
potent anti-IgG and anti-C3. This AGS did not contain tions, cephalothin still binds efficiently to RBCs (in
antialbumin, whereas some commercial AGS did have contrast to penicillin), but the β-lactam ring is much
potent antialbumin. In vitro results indicated that less likely to undergo nucleophilic attack that would
albumin was absorbed readily onto cephalothin- prevent binding of plasma proteins.154 These authors
treated RBCs and could be the cause of false-positive also showed that nonspecific binding of proteins
antiglobulin tests if an AGS rich in antialbumin was decreases as pH decreases. Ninety-seven percent of
used. This could be another reason why Molthan and 133 normal sera reacted, by IAT, with RBCs coated
associates149 or Gralnick and colleagues150 found a with cephalothin at pH 8.5, but only four of 87 (4.6%)
higher percentage of positive DATs than did Spath and reacted weakly with RBCs coated with cephalothin at
coworkers.152 pH 6.0. It was suggested that at low pH, cephalothin
Drug-Induced Immune Hemolytic Anemia 281

CELL

FIGURE 8-10. Nonimmunological adsorption of pro-


teins onto RBCs. Drugs might change the RBC mem-
brane so that many proteins attach to the membrane,
leading to a positive DAT and, possibly, to DIIHA.

Drug

Proteins

might still bind to RBCs (unlike penicillin), but that albumin were found to bind to human RBCs when they
the β-lactam ring would be much less likely to were incubated in vitro with normal plasma and INOX
undergo nucleophilic attack that would permit or with glutaraldehyde, which is similar in structure to
binding of proteins when the RBCs are incubated with INOX. In theory, any drug containing two or more
serum or plasma.14,149 aldehdye groups capable of reacting with proteins can
Garratty and Leger155 were stimulated to defend the produce a positive DAT. These could include the fol-
“membrane modification” concept of Garratty and lowing: actinorhodine, aztreonam, citric acid, digly-
Petz140 because of two reports from 1968.156,157 Sirchia coaldehyde, Evans blue, folic acid, glutamic acid,
and colleagues156 and Ferrone and coworkers157 glutaraldehyde, glutaric acid, glycerol trinitrate (nitro-
showed that cephalothin-treated RBCs acted in some glycerin), glyoxal, iodipamide (Chlorografin), maelic
ways like RBCs from patients with paroxysmal noc- acid, menadiol sodium diphosphate (Synkayite),
turnal hemoglobinuria (PNH). The treated RBCs had pamoic acid, penicillic acid, phthalic acid, picric acid,
enzymatic and metabolic activities similar to those of stibophen, sodium iodomethamate (Iodoxyl), tereph-
PNH RBCs and were similarly sensitive to the action thaldicarboxaldehyde, and terephthalic acid. Jamin and
of complement (e.g., cephalothin-treated RBCs gave a associates160 also reported that suramin (a reverse tran-
positive acidified-serum lysis test [Ham’s Test]). It is scriptase inhibitor that has been used in treating
now known that PNH RBCs have several membrane acquired immunodeficiency syndrome) caused in vitro
abnormalities.158 For instance, phosphatidylinositol- adsorption of IgG onto RBCs associated with positive
glycan (GPI)-anchored proteins, which contain impor- AGTs. No positive DATs have been reported after
tant complement control proteins (CD55 and CD59), suramin administration.
are absent or markedly deficient in PNH RBCs. In Zeger and coworkers161 showed that IgG could be
1995, Garratty and Leger155 confirmed the findings of adsorbed nonimmunologically onto RBCs treated in
Sirchia and colleagues156 and Ferrone and cowork- vitro with cisplatin, an anticancer chemotherapeutic
ers157 and extended them by demonstrating (using agent. They also suggested that a positive DAT due to
flow cytometry) that RBCs treated with cephalothin IgG and complement sensitization, which developed
and cefotetan had markedly decreased quantities of in a patient taking cisplatin, was due to nonimmuno-
RBC membrane CD55 and CD58; CD59 was only logic protein adsorption in vivo. The patient had a
slightly decreased. Thus, the RBC membrane changes hemolytic anemia on presentation, but when rechal-
were different from those demonstrated for PNH lenged with the drug, the patient redeveloped a posi-
RBCs but proved the earlier suggestion by Garratty tive DAT but not a hemolytic anemia. Seven other
and Petz140 that cephalsoporins modify the RBC patients have been reported to have cisplatin-induced
membrane. HA.160-166 Two drugs that are closely related to cis-
platin—carboplatin and oxaliplatin—have also been
Other Drugs Causing Nonimmunologic reported as causing hemolytic anemia.167-172 Oxaliplatin
Uptake of Protein onto RBCs appears to cause positive DATs (and perhaps hemolytic
anemia), due to oxaliplatin antibodies and/or non-
Jamin and associates159 showed that diglycoaldehyde immunologic adsorption of protein onto the RBCs.172
(INOX), an intravenous chemotherapeutic agent used It is interesting that these drugs have been shown to
to treat childhood malignancies, caused positive DATs cause nonspecific binding of protein onto RBCs in vitro,
in all eight patients receiving the drug. IgG and and that there are several cases of hemolytic anemia
282 Immune Hemolytic Anemias

associated with this therapy. Unfortunately, many of Arndt174 also wondered whether Augmentin
these cases had poor serologic evaluations, and fol- (SmithKline Beecham), an antibacterial combination
lowing the findings by Zeger and coworkers,161 it has of the semisynthetic antibiotic amoxicillin and the β-
been assumed that the DATs could have been positive lacatamase inhibitor clavulanate potassium, would
due to nonspecific binding of protein and that the show similar serologic results. They found that the
hemolytic anemia might not have been associated sera from their four patients did not react with RBCs
with this. This assumption was supported by the in the presence of Unasyn or Timentin, but when
knowledge that cephalothin was used commonly for drug-treated RBCs were tested, patients’ sera and
more than 25 years with no cases of HA reported that normal donor sera reacted equally by IAT.174 After
were proved to be due to nonspecific binding of incubation in normal sera, RBCs treated with
protein onto the RBCs. Unasyn, Timentin, Augmentin, sulbactam, and
In 1992, Lutz and Dzik173 reported an analysis of clavulanate reacted by IAT with a range of antiglo-
the cause of all positive DATs at their institution in bulin sera, including antihuman albumin (an index
an 18-month period. They found that 45 of 189 posi- of nonimmunologic adsorption). RBCs treated with
tive DATs (23.8%) were associated with the use of only ampicillin or amoxicillin were nonreactive.174
Unasyn (Roerig Division, Pfizer Inc., New York, NY), Thus, it was concluded that the β-lactamase
an antibacterial combination of the semisynthetic inhibitors sulbactam and clavulanate seem to cause
antibiotic ampicillin sodium and the β-lactamase nonimmunologic adsorption of protein onto RBCs in
inhibitor sulbactam sodium. None of these patients vitro and that this could be the cause of the high inci-
had a HA. They also prospectively followed 59 dence of positive DATs. This study confirmed the
patients with a negative DAT after receiving findings of Williams and associates,175 who in 1985
Unasyn.173 Twenty-three of these patients (39%) had shown that clavulanate could cause in vitro non-
developed a weakly positive DAT (microscopic to 1+ immunologic adsorption of proteins onto human
with an anti-IgG) after a median of 8.5 days of RBCs and was associated with positive DATs in
therapy (range, 3–15 days); there was no associated patients taking drugs containing clavulanate (44% of
hemolytic anemia. patients developed a positive DAT).
These results suggested that Unasyn caused positive Table 8-6 shows the drugs that are thought to cause
DATs far more commonly than any other drug in nonimmunologic uptake of proteins onto RBCs.
present use. Up to that time, the drug most commonly Table 8-7 shows the serologic characteristics associ-
causing positive DATs was methyldopa, with 10–36% ated with drug-induced nonimmunologic adsorp-
of patients developing a positive DAT after 3–6 months tion of proteins onto RBCs.
of therapy, but methyldopa is no longer commonly
used. Upon reviewing their records, Garratty and Nonimmunologic Uptake of Protein
Arndt 174 found that they had studied four patients by RBCs as a Possible Cause of
who developed positive DATs while on Unasyn or a
similar drug, Timentin (a combination of the semisyn- Hemolytic Anemia
thetic antibiotic ticarcillin disodium and the β-lactamase Garratty and Arndt174 further suggested that the
inhibitor clavulanate potassium; SmithKline Beecham nonimunologic uptake of protein (e.g., IgG) might
Inc., Philadelphia, PA). The patients’ sera, or eluates lead to hemolytic anemia. Three of the four patients
from their RBCs, had not reacted with RBCs in the with positive DATs who had received clavulanate or
presence of Unasyn or Timentin, but the patients’ sera sulbactam had an associated hemolytic anemia, and
had reacted with Unasyn- or Timentin-treated RBCs by there was a temporal relationship to the drug
indirect antiglobulin test. As a control, drug-treated therapy. A cause-and-effect relationship could not be
RBCs had always been tested with a pool of normal proven, as no drug antibody was demonstrable.
sera. The pooled normal sera control reacted with the Broadberry and associates176 showed that another β-
drug-reacted RBCs equally well as the patients’ sera, so lactamase inhibitor, tazobactam, could also be associ-
the results with the patients’ sera were initially inter- ated with a positive DAT and HA. They showed that
preted as being insignificant (i.e., no drug antibodies
detected).
The results of Lutz and Dzik173 were reminiscent of
TABLE 8-6. DRUGS ASSOCIATED WITH
earlier reports149,150 of patients receiving cephalothin.
NONIMMUNOLOGICAL ADSORPTION OF
Garratty and Arndt174 wondered whether the reac-
PROTEINS ONTO RBCS
tions they had observed with normal sera and
Unasyn-/Timentin-treated RBCs (results ignored at
cephalosporins
that time) might have been due to in vitro non- cisplatin/oxaliplatin
specific uptake of protein onto the Unasyn- and diglycoaldehyde (INOX)
Timentin-treated red cells, similar to what had been suramin
described with the cephalosporins, and whether this sulbactam (contained in Unasyn)
clavulanate (contained in Augmentin and Timentin)
process might explain the high incidence of positive tazobactam (contained in Zosyn)
DATs observed by Lutz and Dzik.173 Garratty and
Drug-Induced Immune Hemolytic Anemia 283

anemia. We now believe that this could be a new


TABLE 8-7. CHARACTERISTICS ASSOCIATED mechanism for DIIHA.
WITH DRUG-INDUCED NONIMMUNOLOGICAL
ADSORPTION OF PROTEINS ONTO RBCs
SEROLOGIC AND CLINICAL FINDINGS
Patients have positive DAT but often do not have hemolytic anemia. ASSOCIATED WITH DIIHA
Positive DAT might be due to a multitude of proteins on RBC surface
(e.g., IgG, IgM, IgA, C3, albumin).
Nonimmunological adsorption is suggested if the patient’s RBCs The serologic and clinical findings of DIIHA are asso-
react by a DAT using antialbumin (the antialbumin must be ciated with the previous discussion on the mecha-
standardized for use with RBCs). nisms thought to cause DIIHA. Regardless of the
Eluate from patient’s DAT+ RBCs is nonreactive with untreated or
drug-treated RBCs, or in the presence of drug.
theoretic concepts, the serologic and clinical charac-
Patient’s serum may react with drug-coated RBCs, but not with teristics appear to fall into four distinct categories:
untreated RBCs in the presence of drug but normal donor sera
show similar reactions. The strength of reactions might not be 1. The penicillin-type drug-dependent antibodies
similar, as the serum IgG level can influence the nonimmuno- 2. “Nonpenicillin”-type drug-dependent antibodies
logical uptake of protein.
3. Drug-independent (auto-) antibodies
4. Nonimmunologic adsorption of proteins onto RBCs

there appeared to be a relationship with the plasma Most investigators would agree that drug antibodies
IgG level and the positive antiglobulin test. Their can be classified either as drug independent (drug is
patient had a high gamma globulin level; it will be of not needed to demonstrate reactivity, i.e., autoanti-
interest, therefore, to attempt to correlate plasma IgG bodies) or as drug dependent (presence of drug is
levels in future patients with hemolytic anemia sus- needed to demonstrate reactivity). We prefer to
pected to be due to this mechanism. Arndt and categorize the latter group into “penicillin-type” and
Garratty177 provided further data to support their “nonpencillin-type” antibodies. We do this because
hypothesis by showing that after in vitro incubation the serologic and clinical characteristics of these
with clavulanate, sulbactam, or tazobactam, normal groups are so different (see Tables 8-4 and 8-5).
RBCs were phagocytosed by monocytes in a mono- The penicillin-type drug-dependent antibody was
cyte monolayer assay (MMA). This MMA has been said to react by a “drug adsorption” mechanism,
pedigreed as an assay to predict in vivo RBC sur- implying that drug is adsorbed onto the RBC mem-
vival.178,179 The MMA results were strongly positive, brane and the drug antibody reacts with the drug-
suggesting that RBCs coated with proteins by coated RBC. We see nothing wrong with continuing
nonimmunologic means could have shortened RBC this terminology but could readily accept an alterna-
survival. tive descriptive term to describe the mechanism. We
As discussed previously, the most convincing evi- do not like the term “hapten mechanism” because it
dence to prove that a drug is the cause of a hemolytic could be applied to all drugs.
anemia is to show that hemolytic anemia develops Whether we should continue to use the term
when the patient receives the drug again (accidentally “immune complex” mechanism to explain how
or deliberately). This approach is obviously not drug-dependent antibodies other than pencillin react
usually possible, although it has been used to prove or is open to debate. Some workers feel strongly that
disprove the role of the drug in the hemolytic because of the recent criticisms of the theory, we
process.9,10,39,141 Although it has been argued that this should not continue to use the term. We have an
is the most definitive proof to incriminate the drug in intermediate view and feel that as no other theory
question, some of the early methyldopa data are of has been proven to be correct, it is acceptable to use
interest. Breckenridge and coworkers9 reintroduced such terms as “the so-called immune complex mecha-
methyldopa in three patients whose methyldopa- nism,” or to put the words “immune complex” in
induced positive DATs had become negative and quotation marks to emphasize that it is not a proven
found that only one of the three developed a positive fact. It is, of course, perfectly acceptable to use the
DAT again. It is interesting to find, on once again terms immune complex “hypothesis”or “theory”
reviewing the cisplatin publications, that six of the without adulteration. It is difficult for us to com-
eight patients with cisplatin-associated hemolytic pletely reject that drug immune complexes some-
anemia were rechallenged with cisplatin following times form, as the serologic findings (e.g., very small
remission of the positive DATs and hemolytic amount of drug necessary for in vitro reactions; com-
anemia.161-166 DATs became positive again in all cases, plement activation leading to lysis of RBCs in vitro)
and three developed hemolytic anemia again. These and clinical findings (acute intravascular hemolysis
data, together with the data reported in the foregoing and sometimes renal failure following ingestion of
discussion, cause us to question our previous assump- very small quantities of drug) are so typical of
tions that cisplatin (and other drugs that appear to immune complex–mediated reactions. We like many
cause only nonspecific uptake of proteins onto RBC aspects of the unifying hypothesis, however.118,119
membranes) cause positive DATs but not hemolytic Perhaps Shulman and Reid’s139 suggestions are a
284 Immune Hemolytic Anemias

happy compromise because they appear to embrace 100 DAT


the best of both approaches. 75 (score)

50
Drug-Dependent Antibodies 25
Drug-dependent antibodies appear to fall into two 0
groups serologically: those that react with drug-coated
RBCs (see Table 8-2), and those that are detected only 12
Hemoglobin
when drugs, RBCs, and antibody are mixed. Of the 11

gm/100 mL
drugs listed in Table 8-2, only the penicillins and some- 10
times cephalothin present with consistent characteris- 9
tics that are different from most other drugs. Among
patients receiving large doses of these drugs intra- 8
venously, 3–5% develop a positive DAT, and a small 7 Transfusions
percentage of these develop IHA associated with IgG-
15
mediated extravascular hemolysis. In contrast, the Reticulocytes

Percent
other drug-dependent drugs are usually associated 10
with clinical symptoms more typical of immune- 5
complex mediated disease (i.e., acute complement- 0
mediated intravascular hemolysis, sometimes
Quinidine
associated with renal failure). Streptomycin
Penicillin
PENICILLIN ANTIBODIES
26 31 10 15 20 25 5 11 16 21 26 31
1/21 2/5 3/1
Between 1959 and 1965, four patients were reported as
having hemolytic anemia during penicillin administra- FIGURE 8-11. Course of a patient who developed immune hemolytic
tion, and also as having a positive DAT and penicillin anemia while receiving intravenous penicillin for suspected bacterial
endocarditis. The direct antiglobulin (Coombs’) test was known to have
antibodies in their sera. All these patients had serious been negative prior to initiation of penicillin therapy. (From Petz LD,
systemic infection, however, and they also were receiv- Fudenberg HH: Coombs-positive hemolytic anemia caused by penicillin
ing other drugs. For instance, Ley and colleagues180 administration. N Engl J Med 1966;274:171–178.)
described a patient receiving 18 million units of peni-
cillin daily who was found to have a positive DAT (no
prior negative DATs had been recorded). On the was challenged with penicillin at a time when there
seventh day of penicillin therapy, probenecid was was no evidence of infection, to clarify the role of
added to the regimen, and at that time the hematocrit penicillin in production of hemolysis. All the original
level began to fall. Despite blood transfusion the fall findings were reproduced (Fig. 8-12), thereby docu-
continued. Therapy with both penicillin and probe- menting the etiologic role of penicillin.
necid was then stopped; after further transfusions, the Further observations indicated that penicillin dif-
hematocrit level remained stable. The patient subse- fered from drugs that had previously been described
quently died; penicillin antibody was demonstrated in as causing thrombocytopenia or hemolytic anemia in
a sample of blood drawn on the day of death. that it was strongly bound to the RBC membrane in
In 1966, Petz and Fudenberg141 were the first to vitro in the absence of antibody. Indeed, when a
systematically study a case of penicillin-induced serum containing penicillin antibody was tested
hemolytic anemia; they described a patient who against RBCs coated with penicillin, there was no
developed hemolytic anemia during therapy with decrease in titer even after the RBCs were washed
high doses of penicillin for suspected bacterial endo- 20–25 times with buffered saline; studies with tritium-
carditis (Fig. 8-11). Serologic tests revealed a strongly labeled penicillin also indicated that penicillin was
positive DAT with anti-IgG but negative reactions “firmly bound” to the RBCs.141
using an anti-“nongamma” reagent. In spite of the In approximately 3% of the patients who receive
strongly positive DAT, no antibodies could be massive doses of intravenous penicillin, positive
detected in the patient’s serum or in an eluate from DATs will develop, but only a small percentage of
the patient’s RBCs using standard serologic tech- these develop an obvious hemolytic anemia.142,181
niques. Using penicillin-coated RBCs, however, peni- Levine182 reported that 5–10% of patients receiving
cillin antibody was demonstrable in the serum by long-term, high-dose penicillin could develop mild
direct agglutination, by IAT, and in an eluate from the hemolytic anemia that might not be noticed because
patient’s RBCs. After the cessation of all drugs except of the underlying disease. The mechanism of the posi-
prednisone, hemolysis persisted for about 3 weeks tive DAT and hemolytic anemia seems clear.1,141 The
and then gradually resolved. The DAT became pro- drug is adsorbed to the RBCs, and the penicillin anti-
gressively weaker and was negative 6 weeks after bodies present in the patient’s plasma react with the
cessation of the drug. Seven months later, the patient penicillin on the RBCs (see Fig. 8-9). The quantity of
Drug-Induced Immune Hemolytic Anemia 285

mg/100 mL
150 who had intravascular hemolysis of life-threatening
100 severity with hemoglobinemia and hemoglobinuria
Haptoglobin
50 (Fig. 8-13).147 Significant amounts of complement
0 components C3 and C4 were detected on the
patient’s RBCs, in addition to the usual IgG antibody
15 to penicillin. The patient’s serum caused direct
Hemoglobin agglutination of penicillin-coated RBCs and reacted
gm/100 mL

13 by IAT using anti-IgG, but not anti-C3. The peni-


11 cillin-coated RBCs were not lysed in vitro, even in
the presence of fresh complement. Complement
9 fixation could be demonstrated, however, by incu-
bating the patient’s serum with normal serum and
then measuring residual hemolytic complement
Percent

10 Reticulocytes activity. Similar complement fixation could also be


5
0 demonstrated using serum from a patient with peni-
cillin-induced serum sickness, but fixation could not
be demonstrated with the serum of a patient with
Millions of
units daily

100
50 Penicillin
penicillin-induced hemolytic anemia without
0 intravascular hemolysis, or with the sera of three
people with relatively high titers of antibody to peni-
Capa titer cillin but without hemolytic anemia. The presence of
1:256 complement components on the patient’s RBCs and
1:64 the finding that her serum fixed complement in vitro
1:16 suggested that penicillin-antipenicillin immune com-
1:4 plexes were present in her serum. We further sug-
gested that RBC damage mediated by immune
10 complexes might have augmented the noncomple-
4 Fold dilutions

8 DAT (titer)
ment, IgG-mediated hemolysis that is usually seen
of AGS

6
4 in penicillin-induced IHA.147 The unusual severity
2 of hemolysis appeared to result from the high titer of
0 antibody to penicillin (8000) and to participation of
20 30 10 20 30 9 19 29 8 18 the complement system in hemolysis. Funicella and
9/15 10/5 11/4 12/3
coworkers148 reported a patient who was treated
FIGURE 8-12. Documentation of etiologic role of penicillin in causation with ampicillin, oxacillin, and penicillin over an 11-
of immune hemolytic anemia. The patient is the same as in Figure 8-11
and a repeated administration of penicillin caused recurrence of all the
day period. Hemolytic anemia ensued, and the DAT
original findings. (From Petz LD, Fudenberg HH: Coombs-positive was positive with both anti-lgG and anti-C4/C3. The
hemolytic anemia caused by penicillin administration. N Engl J Med authors demonstrated the presence of immune com-
1966;274:171–178.) plexes using a radiolabeled Clq-binding test, and
they suggested that RBC destruction occurred both
by the immune complex mechanism and the drug
penicillin (BPO) antibody sensitizing the RBC is absorption mechanism (which they referred to as the
limited by the number of BPO haptenic groups on the “haptene type”).
cell, the plasma concentration of BPO-specific anti- Our findings in 10 patients with penicillin-induced
bodies, and the avidity of the antibodies. Intravascular hemolytic anemia (which includes the patient with
hemolysis rarely occurs; IgG-sensitized RBCs are intravascular hemolysis described previously) indicate
removed extravascularly by the reticuloendothelial that in a majority of patients, the DAT is positive only
system in the same way as Rh (IgG) sensitized cells with anti-IgG.1 Four patients in our series and several
(see Chapter 4). It is interesting to note that although patients reported by others, however, also had comple-
intravascular lysis rarely occurs, penicillin antibodies ment components on their RBCs.1 Although these
often hemolyze penicillin-coated RBCs in vitro (the numbers are small, they suggest that hemolysis might
hemolytic antibodies are always of low titer), be augmented by complement activation in a significant
and complement is detectable on 40% of the RBCs number of patients. This process might not be efficient
from patients with a positive DAT due to penicillin enough to cause intravascular hemolysis but could
antibodies.1 enhance extravascular RBC destruction within the reti-
Kerr and colleagues146 described a patient whose culoendothelial system (RES) (see Chapter 4).
RBCs reacted strongly with both anti-IgG and anti- One should appreciate that a positive DAT occur-
complement antiglobulin sera, and they suggested ring during penicillin administration is not, in itself,
that complement activation could contribute to an indication for cessation of the drug. Approximately
immune hemolysis in some cases. In addition, we 3% of patients receiving high doses of intravenous
described a patient with penicillin-induced IHA penicillin develop a positive DAT, but only a small
286 Immune Hemolytic Anemias

50%

Reticulocytes

Hematocrit
40%

30% FIGURE 8-13. Clinical course of a patient with peni-


cillin-induced hemolytic anemia with intravascular
hemolysis of life-threatening severity. The patient’s
– serum contained penicillin antibody to a titer of
8000, and her RBCs were sensitized with IgG, C3,
and C4. She refused transfusion because of
religious beliefs and was treated with oxygen,
20% parenteral fluids, and corticosteroids. (From Reis
CA, Rosenbaum TJ, Garratty G, et al: Penicillin-
induced immune hemolytic anemia. JAMA
1975;233:432–435.)

10%
Penicillin therapy,
40 million units 2 gm
daily intravenously daily orally
Cephalothin,
2 pm daily
intravenously orally
Dexamethasone,
Probenecid, 2 gm daily orally
40 mg daily (tapared)

0 4 8 12 16 20 24 28 32 36
Hospital day

percentage of these patients develop hemolytic caused positive DATs and IHA. Usually, the anti-
anemia. Hemolysis might not ensue, even with con- bodies detected in the patients’ sera react with RBCs
tinued administration of the drug. coated with the putative penicillins (e.g., ampicillin,
There is no direct correlation between the presence of amoxicillin, piperacillin, and ticarcillin) and cross-
IgG and IgM penicillin hemagglutinating antibodies react with penicillin-coated RBCs.188-194 On rare occa-
and allergic reactions, but high-titer IgG antibodies sions, this cross-reaction is not observed. Kroovand
were found to occur more often in the allergic group.182 and associates195 and Garratty and Brunt196 reported
The clinical and laboratory features of penicillin- on two patients who had positive DATs caused by naf-
induced IHA are quite constant and are listed in cillin antibodies. The antibodies reacted with nafcillin-
Table 8-4. coated RBCs but not with penicillin-coated RBCs. The
Some of the cephalosporins (e.g., cephalothin and antibodies could be adsorbed with nafcillin- and
cefamandole) also caused drug-induced positive methicillin-coated RBCs but not with penicillin-
DATs and IHA by a mechanism that appeared the coated RBCs. Nafcillin and methicillin are the only
same as that for penicillin.152,183-187 All of the patients penicillins that lack a CH group between the side
with hemolytic anemia had RBCs sensitized with IgG chain and the β-lactam ring. Garratty and Brunt196
antibodies to the appropriate cephalosporin, and their suggested that the CH group in penicillin could steri-
sera reacted with cephalosporin-coated RBCs; no cally interfere with interaction of the nafcillin anti-
intravascular hemolysis was observed. These results bodies. This does not always occur, because we have
were in contrast to the more dramatic complement- encountered nafcillin and methicillin antibodies that
mediated intravascular hemolysis associated with react well with penicillin-coated RBCs.
some of the newer second- and third-generation On occasion, some penicillins yield unexpected
cephalosporins (to be discussed shortly). serology. Arndt and coworkers192 described two
patients with cystic fibrosis who developed positive
DIIHA ASSOCIATED WITH PENICILLINS DATs and hemolytic anemia after 11–12 days of
OTHER THAN PENICILLIN G therapy with piperacillin or Zosyn (which contains
piperacillin) and a β-lactamase inhibitor, tazobactam
Penicillins (including semisynthetic products), other sodium. Both patients’ sera were shown to contain
than penicillin G, that are used intravenously have piperacillin antibodies. One patient’s serum contained
Drug-Induced Immune Hemolytic Anemia 287

an IgG complement activating antipiperacillin that did


not react with piperacillin-coated RBCs but did react TABLE 8-8. DRUGS THAT HAVE BEEN REPORTED
with untreated RBCs in the presence of piperacillin (i.e., TO INDUCE RBC DRUG-INDEPENDENT ANTIBODIES
“immune complex” method). The second patient’s (I.E., AUTOANTIBODIES)
serum contained IgG + IgM complement-activating
Group Ia Group IIc
piperacillin antibodies that agglutinated piperacillin-
coated RBCs and reacted with untreated RBCs in the cladrabine azapropazone
presence of piperacillin. The latter patient died of fludarabine carbimazole
severe intravascular hemolysis. levodopa ceftazidime
mefenamic acid cefoxitin
methyldopa cefotetan
DRUG-DEPENDENT ANTIBODIES OTHER procainamide cefotaxime
THAN “PENICILLIN TYPE” (“IMMUNE catergenb chlorinated hydrocarbons
chaparralb diclofenac
COMPLEX” MECHANISM) chlordiazepoxideb glafenine
cianidanol latamoxef
This group of drug-dependent antibodies, which cyclofenilb nomifensine
often is classified as of the “immune complex” mech- diphenylhydantoinb phenacetin
anism type, includes the largest number of drugs, but fenfluradimeb streptomycin
many of them are represented by a single case report ibuprofenb teniposide
interleukin-2b tolmetin
in the literature. Table 8-5 shows the clinical and sero- methoinb zomepirac
logic characteristics of this group. methysergideb
The group is characterized by acute intravascular rituximabb
hemolysis, sometimes associated with renal failure. a These drugs have been reported to induce drug-independent antibodies only.
The patient’s RBCs are often sensitized with only com- b More evidence is needed to prove that these drugs really can induce RBC
plement components (e.g., C3dg), but IgG is some- autoantibodies.
c These drugs induce drug-independent antibodies together with drug
times also present; on rare occasions only IgG is
dependent antibodies reacting by different mechanisms (see Table 8-3).
detected. The patient’s serum reacts with RBCs in the
presence of the drug and/or of a metabolite of the
drug. Using drugs from this group, it is usually not
possible to prepare drug-coated RBCs without using good example of this is the cimetidine story discussed
chemical coupling techniques (e.g., cross-linking earlier in this chapter. Unfortunately, it is extremely
reagents); thus, RBCs pretreated with drug do not difficult to perform such important confirmatory in
react with the patient’s serum.197,198 The drug- vivo experiments as those performed by Petz and
dependent antibodies could be IgG and/or IgM; they colleagues,10 and such experiements could, in fact, be
almost always activate complement. In the presence dangerous for a patient whose initial symptoms
of the drug, they can cause lysis, agglutination, and/ included acute intravascular hemolysis rather than
or sensitization of RBCs detectable by the antiglobulin only a positive DAT or milder extravascular RBC
test. Enzyme-treated RBCs almost always react more destruction.
strongly than untreated RBCs. Table 8-8 divides the drugs into two groups: those
that induce drug-independent antibodies alone and
Drug-Independent Antibodies those that induce drug-independent antibodies
together with drug-dependent antibodies. We believe
Drug-independent antibodies appear as autoantibo- that autoantibody production in the two groups
dies by serologic testing. That is to say, the antibodies could be induced by different mechanisms. The most
are proven to be induced by a drug but react in vitro thoroughly investigated drugs known to induce true
without drug being present. Table 8-8 lists drugs that autoantibodies are methyldopa and procainamide.
have been reported to cause autoantibody production. In a 10-year period (1970–1980) studying immune
It is difficult to prove that a particular drug is respon- hemolytic anemias, we reported that almost 70% of
sible for appearance of autoantibodies. As the anti- the DIIHAs we encountered were due to methyldopa
bodies react in vitro with RBCs in a way identical to (23% were due to penicillin).1 Methyldopa was not
non–drug-induced autoantibodies, they cannot be dif- used as much in the 1990s, and there are no reports
ferentiated serologically. It is not acceptable to blame on a large series similar to the one we reported in
a drug just because a patient forms RBC autoantibo- 19801 that would reflect on the incidence of methyl-
dies after drug administration; this could be coinci- dopa-induced hemolytic anemia in the 1990s. Suffice
dental, and often is. If a patient forms autoantibodies it to say that we have not had a case of methyldopa-
after drug administration and hemolysis resolves after induced hemolytic anemia referred to us in the last
discontinuation of the drug, the evidence is better; but 10 years. Most of the cases of drug-induced AIHAs
it still does not prove the drug caused the hemolysis; are associated with fludarabine (see the discussion
the two events could still be coincidental. Often, the later in the chapter). Nevertheless, it is worth dis-
patient is also treated with steroids, and remission cussing the characteristics of methyldopa-induced
might not be due to discontinuation of the drug. A hemolytic anemia, as it is the prototype of AIHA
288 Immune Hemolytic Anemias

associated with drugs and has been investigated more those without hemolytic anemia, they were unable to
thoroughly than other drugs. Table 8-9 lists the char- select a distinct “hemolytic threshold” to differentiate
acteristics associated with methyldopa-induced RBC these groups. The range of RBC-bound IgG showed
autoantibodies. considerable overlap in each group of patients studied.
There are two unanswered questions regarding The differences between the conclusions of van der
methyldopa-induced autoantibodies: Meulen and coworkers199 and Garratty and Nance200
could not be explained by the subclass of the RBC-
1. Why do up to 15% of patients receiving methyldopa bound IgG. Garratty and Nance200 studied a larger
make autoantibodies to their own RBCs? number of patients (104 vs. 29) covering a more
2. Why do only 0.5% of patients with a positive DAT diverse population of hemolytic anemias; 12 of the
due to methyldopa have hemolytic anemia? 17 cases of hemolytic anemia studied by van der
Meulen and coworkers199 were due to methyldopa,
van der Meulen and coworkers199 suggested the exist- whereas the series of 28 patients with hemolytic
ence of an in vivo hemolytic quantitative threshold. anemia reported by Garratty and Nance200 included 7
They used flow cytofluorometry to study 29 patients idiopathic cases, 8 methyldopa-induced cases, and 13
with positive DATs due to IgG1 autoantibodies; 17 of with hemolytic disease of the newborn. The conclu-
the patients had signs of overt hemolysis, and 12 had no sions of Garratty and Nance200 that no definitive
obvious hemolysis. Twelve of the patients were receiv- “hemolytic threshold” existed, were the same when
ing methyldopa; seven of these had overt hemolysis, the methyldopa-induced group was analyzed sepa-
while the other five had no hemolytic anemia. The rately from the total group of hemolytic anemias. In
authors were able to show a distinct difference in the conclusion, although we believe that there is good
number of IgG molecules on the RBCs of patients with evidence to suggest that the amount of RBC-bound
and without AIHA. There appeared to be a “thresh- IgG is a major factor in determining the degree of in
old”—a critical degree of sensitization—above which vivo RBC destruction, there are significant discrepan-
increased RBC destruction in vivo became apparent. cies between the amount of RBC-bound IgG and the
Only two discrepant results were found, one in each degree of in vivo hemolysis seen in many cases.
group of patients; neither of these were patients taking
methyldopa. van der Meulen and coworkers199 con- Laboratory and Clinical Findings
cluded that the quantity of IgG1 autoantibody on the
RBCs of DAT-positive patients taking methyldopa was
Associated with Methyldopa
the determining factor for in vivo hemolysis to occur. Administration
Garratty and Nance200 also used flow cytometry to LABORATORY FINDINGS
study 104 individuals with positive DATs, with and
without AIHA, but they were unable to confirm this Direct Antiglobulin Test. Carstairs and associates201
conclusion. They confirmed that flow cytometry was performed serologic studies on 202 consecutive
much better than the AGT (i.e., titration scores) for dif- hypertensive patients receiving methyldopa and com-
ferentiating RBCs with different amounts of IgG on pared the results with those in 76 control patients who
them, particularly when they were strongly sensitized had never received methyldopa. Forty-one of the 202
(3+ to –4+). Although their results confirmed that the patients (20%) had positive DATs, all of which were
mean amount of RBC-bound IgG was always higher in caused by coating of the RBCs with IgG only. None of
patients with hemolytic anemia due to autoantibodies the control patients had a positive DAT of the pure
(idiopathic and methyldopa-induced) or alloantibodies IgG type, but two had a positive test of non-IgG type.
(hemolytic disease of the newborn), compared with Of 65 patients taking 1 g methyldopa or less per day,
7 (11%) had a positive DAT; of 86 patients taking 1–2
g per day, 16 (19%) had a positive test; and of 51
patients taking more than 2 g per day, 18 (36%) had a
TABLE 8-9. CHARACTERISTICS ASSOCIATED WITH positive DAT.
METHYLDOPA-INDUCED RBC AUTOANTIBODIES The development of a positive DAT usually occurs
only after 3–6 months of treatment with the drug,
10%–30% of patients taking methyldopa develop RBC autoantibo-
dies (i.e., positive DAT) within three to six months of therapy.
although Hunter and colleagues202 found that 11 of 26
Incidence of positive DAT is dose dependent. patients with a positive DAT gave a positive reaction
Only about 0.5% of patients develop hemolytic anemia. only after 3 or more years of therapy. This delay is not
Serological findings are similar to those associated with idiopathic shortened when a patient who previously gave a posi-
AIHA, particularly those with only IgG on their RBCs: tive test is restarted on the drug.201,203-205
IgG autoantibody on RBCs (17% have weak C3 sensitization in
addition to IgG) The RBCs of patients with no evidence of hemolysis
IgG autoantibody often present in serum show varying degrees of reactivity in the DAT, whereas
Usually “Rh” specificity in patients who have overt hemolysis, the reaction is
Following cessation of drug therapy, the hemolytic anemia resolves characteristically strongly positive.1,6,205 In a series of
quickly (usually within two weeks), but DAT might remain positive
for up to two years.
29 patients with methyldopa-induced hemolysis, the
DAT was 21⁄2+ to 4+ in all cases, with 66% of the
Drug-Induced Immune Hemolytic Anemia 289

patients having a 31⁄2+ to 4+ reaction. Worlledge205 methyldopa in concentrations of 0.1 to 10 mmol/L to


reported that all patients with methyldopa-induced sera or eluates, but this did not inhibit the activity of
AIHA had a negative DAT using anti-C3, although we eluates toward normal RBCs or of serum activity
found that in four of 29 (14%) patients, the DAT was toward enzyme-treated cells. The following metabo-
also weakly positive with anti-C3.1 lites or congeners of methyldopa yielded similar results:
Most patients taking methyldopa have little or no
evidence of a shortened RBC survival. The incidence • 1-(3,4-dihydroxyphenyl)-2-amino propanol
of hemolysis depends on the criteria used for the diag- • D-2 methyl-3,4-dihydroxyphenylalanine hydrate
nosis, and minor degrees of hemolysis are probably • L-2 methyl-3,4-dihydroxyphenylalanine hydrate
not infrequent.206 Six of the 202 patients taking • R(-) l-(3,4-dihydroxylphenyl)-2-amino propane
methyldopa who were studied by Carstairs and hydrochloride
colleagues201 had reticulocyte counts between 3 and • l-(3-methoxy-4-hydroxyphenyl)-2-amino propane
6%, and one of four RBC survival studies performed hydrochloride
on patients with a positive DAT showed a survival • l-(3-methoxy-4-hydroxyphenyl)-2-propanone
slightly less than normal. In a series of 572 patients on • (+)4-(2-amino propyl)-2-methoxyphenol hydro-
methyldopa therapy, two patients (0.3%) developed chloride
overt hemolytic anemia.206 In a review of 1395 patients
receiving methyldopa, 0.8% had overt hemolysis.6 The results of inhibition and enhancement experi-
Characteristics of the Autoantibody. IgG antibody ments were also reported by Worlledge.108 They
can be eluted from the RBCs of patients with methyl- attempted to inhibit the antibodies in patients’ sera
dopa-induced IHA and has been shown to be an by incubation with methyldopa or its derivatives,
autoantibody that reacts with normal RBCs without including methyldopa hydrochloride, D-α-methyl-
the addition of the drug.206 The eluate can be reactive dopamine hydrochloride, L-α-methyldopamine
even in patients who have a positive DAT but do not hydrochloride, and L-α-methyl-noradrenaline. Sera
have AIHA. Bakemeier and Leddy,207 in a detailed taken from three patients 3 hours after ingestion of
study of one patient, showed that the eluted antibod- 500 mg of methyldopa tablets was also tested as an
ies, although all IgG in type, had both kappa and inhibitor.
lambda light chains and were of three distinct heavy Additional experiments were performed in an
chain subclasses. These findings are similar to those in attempt to enhance the activity of the serum autoanti-
idiopathic AIHA. body by using RBCs that had previously been incu-
Lalezari and colleagues208 used an antiglobulin test bated with methyldopa, its derivatives, or the serum
potentiated by polyvinylpyrrolidone (PVP) and of patients on methyldopa therapy.
detected IgG, IgM, and the first component of com- None of the in vitro tests by LoBuglio and Jandl209
plement (Clq) on erythrocytes of eight patients who or by Worlledge and colleagues105 yielded positive
had hemolysis associated with methyldopa adminis- results; both groups of investigators concluded that
tration, but they found only IgG on the RBCs of three by none of the techniques available could it be estab-
patients who did not have hemolysis. When the lished that methyldopa participated as antigen or
hemolyzing patients recovered, IgM and Clq were no hapten in sensitizing RBCs.
longer detectable. These investigators proposed that Specificity of Drug-Induced Autoantibodies.
high-affinity warm-reactive IgM antibodies (not the Worlledge205 tested eluates from RBCs of 26 patients
IgG antibodies) were primarily responsible for hemol- with methyldopa-induced hemolysis against two
ysis. IgG and its subclasses were variably present on samples of Rhnull RBCs (one with abnormalities of the
RBCs of all patients, regardless of hemolytic activity. S, s, and U antigens and one without these abnormali-
Unfortunately, this finding has not been studied by ties), two samples of U-negative RBCs, one sample of
other investigators. LW-negative RBCs, and RBCs of ordinary Rh geno-
Attempts to Demonstrate Specificity for Drug- types. One third of the eluates showed some specificity
Related Antigens. LoBuglio and Jandl209 attempted to for cells of ordinary Rh genotypes on dilution, and all
demonstrate specificity of the antibody for drug- but two eluates either did not react or yielded very
related antigens by several methods. They incubated weak reactions with both samples of Rhnull RBCs while
normal RBCs with methyldopa in concentrations of reacting strongly with the other cells. The two eluates
1–5 mmol/L for 2 hours at 37°C and for 18 hours at 4°C; that seemed to react as well with Rhnull RBCs as with
after subsequent incubation with the patients’ sera, no other RBCs were adsorbed with all the RBC samples.
agglutination or sensitization to antiglobulin serum Adsorption with the Rhnull RBCs left an antibody that
took place. At higher concentrations of drug, serum reacted only with the other RBCs and did not react with
proteins adsorbed nonspecifically to the RBCs, and the Rhnull RBCs; adsorption with other RBCs removed
there was no difference between patients’ and control all of the antibody. None of the eluates showed any
sera. Also, the mixture of methyldopa in concentrations anti-U or anti-LW specificity, and the antibodies in the
of 1–5 mmol/L added to mixtures of normal RBCs and serum reacted in a similar way.
patients’ sera did not induce a positive antiglobulin Specificity for Rh antigens was confirmed by other
reaction. Inhibition tests were performed by adding investigators.207,209 Bakemeier and Leddy207 showed
290 Immune Hemolytic Anemias

that at least four serologically separable antibodies 100


were present: anti-c, anti-e, and two antibodies react- 90
ing with two defined antigens or groups of antigens 80
absent from Rhnull RBCs. The most common specifi-
city of autoantibody is against antigens of the Rh 70

Percent sensitized cells remaining in the circulation


system. Much less commonly reported target antigens 60
are Wrb, Jka, and U.210-212 The proportion of cases in
which the antibody is directed against an antigen 50
within the Rh system varies from 100% to 30%.204,207 In
some cases, the antibody has specificity for more than
40
one antigen.1,207
Thus, it is remarkable that the antibodies not only
react with normal RBCs but also show Rh specificity,
30
and the pattern of specificity is similar to that of
autoantibodies eluted from the RBCs or present in the
serum of patients suffering from “idiopathic” AIHA of
warm antibody type (see Chapter 7).
Reticuloendothelial Function. Branch and cowork- 20
ers213 and Gallagher and associates214 studied mono-
cytes and macrophages collected from the peripheral
blood of patients receiving methyldopa and found DAT +
No hemolysis { DAT –
that the circulating monocytes of the hemolyzing
patients (but not of the nonhemolyzing patients) Hemolysis DAT +
phagocytosed the DAT positive RBCs in vitro. Yust
and colleagues215 have reported similar findings.
Other investigators have studied reticuloendothelial 10 20 30 60
function in vivo by measuring the rate of clearance of Time (minutes)
radiolabeled autologous RBCs that have been heat FIGURE 8-14. The clearence of IgG-sensitized Cr-labeled autologous
damaged or sensitized by alloantibodies. Kelton216 RBCs in patients taking methyldopa. Four patients had positive DATs
studied reticuloendothelial function in nine patients and no hemolysis (•—•), and four patients had negative DATs and no
taking methyldopa. Five of the nine patients had a hemolysis (°---°). One patient had a positive DAT and hemolytic anemia
positive DAT. Only one patient had laboratory evi- (•---•). The hatched area represents the mean (±SD) clearance in 12
healthy controls. (Reprinted by permission of Kelton JG: Impaired
dence of hemolysis. The patients without hemolysis reticuloendothelial function in patients treated with methyldopa. N Engl
had significantly impaired reticuloendothelial clear- J Med 1985;313:596–600.)
ance. In contrast, the patient with hemolysis did not
have impaired reticuloendothelial function. Kelton216
suggested that the absence of hemolysis in many chromasia and were indistinguishable from the films of
patients with methyldopa-induced autoantibodies other cases of AIHA of the warm antibody type.
could be caused by an impairment in reticuloendothe-
lial function and, moreover, that the drug itself might
be responsible for the impairment because abnormal DIAGNOSIS OF DRUG-INDUCED AIHA
reticuloendothelial function was found in patients
taking the drug who had a negative DAT (Fig. 8-14). There are no diagnostic serologic features of drug-
induced AIHA that distinguish such cases from AIHA
CLINICAL FEATURES not caused by a drug. Thus, the diagnosis rests on the
demonstration of characteristic serologic findings of
Hemolytic anemia has been diagnosed as early as AIHA of warm antibody type in a patient taking a
18 weeks and as late as 4 years after the start of treat- drug known to cause AIHA. Resolution of hemolysis
ment with methyldopa.217,218 Even though the inci- after discontinuing the drug strengthens the diagno-
dence of a positive DAT increases with higher doses of sis, and recurrence of immunohematologic findings
the drug, there is no relation between the amount of and hemolysis after restarting the drug confirms the
methyldopa taken and the intensity of the anemia.201 diagnosis, but taking this step is ordinarily not appro-
Eighteen of the 25 patients reported by Worlledge and priate. Although there are no diagnostic laboratory
colleagues105 were taking 1 g or less of methyldopa per features, certain findings are particularly characteris-
day. In some patients, the onset of hemolysis seemed to tic of methyldopa-induced AIHA and, if absent,
be acute, but in most it was insidious. would tend to exclude the diagnosis. If the DAT is
In 13 of the 25 cases (52%), enlargement of the liver negative or only weakly positive using anti-IgG or if
and lymph nodes was absent but a variable degree of no autoantibody is detected in the patient’s serum, the
splenomegaly was present. In all anemic patients, the diagnosis of methyldopa-induced AIHA is highly
peripheral blood films showed spherocytosis and poly- unlikely.1,105,213
Drug-Induced Immune Hemolytic Anemia 291

BLOOD TRANSFUSION ing that 15 of 19 patients (79%) rechallenged with


fludarabine experienced recurrence of the AIHA.
The indications for blood transfusion and the methods Convincing data also came from a prospective
appropriate for compatibility testing in patients with randomized trial of cyclophosphamide, doxorubicin,
methyldopa-induced AIHA are the same as those for and prednisone (CAP) vs. fludarabine in newly
warm antibody AIHA not associated with methyldopa diagnosed or relapsed CLL patients.243 In the fludara-
administration (see Chapter 10). Some patients who bine arm, 5% of the patients had autoimmune phe-
have a positive DAT caused by methyldopa but who nomena (2% AIHA, 2% ITP, 1% pure red cell aplasia),
do not have evidence of hemolysis might nevertheless whereas no patients in the CAP arm had autoimmune
have a positive IAT. Also, Worlledge217 reported that a phenomena.243
positive DAT is always accompanied by free IgG Di Raimondo and coworkers224 found only five
autoantibodies demonstrable with enzyme-treated cases of AIHA among 112 CLL patients without pre-
RBCs. The presence of autoantibody in the patient’s existing HA who were treated with fludarabine; a
serum, especially when reactive by IAT, results in an further four patients had preexisting HA that deterio-
incompatible cross-match test and makes difficult the rated following treatment with fludarabine. Vick and
detection of RBC alloantibodies. This problem can be associates232 have described a case of “mixed-type”
circumvented by adsorbing the autoantibody from the AIHA following the fifth cycle of fludarabine therapy.
serum using RBCs that do not adsorb alloantibodies Myint and colleagues226 reported that hemolysis
from the patient’s serum, that is, by performance of occurred after a mean of four courses of fludarabine
warm autoadsorption tests using autologous RBC or and that the hemolysis tended to be severe and to
selected allogeneic RBC (see Chapter 10). It is reassur- have an abrupt onset. The HA responded to steroid
ing that, if no alloantibodies are present and the therapy (1 mg/kg). In some cases, it was possible to
patient has no laboratory evidence of hemolysis, trans- administer further fludarabine safely, but commonly,
fused RBCs seem to survive normally even when both further courses exacerbated the hemolysis.
the DAT and IAT are positive.219 Autologous blood for Some reports describe catastrophic hemolysis
transfusion in patients who have a positive DAT and leading to a fatal outcome.227,228 Such massive hemo-
IAT but who do not have AIHA also may be used.220,221 lysis appearing abruptly is not characteristic of AIHA
Some patients receiving methyldopa and in need of occurring in CLL and is further evidence of the effect
blood transfusion might have a positive DAT but a of fludarabine. An additional case of fatal AIHA was
negative IAT and no evidence of hemolysis. For these reported by Byrd and coworkers225 in a patient who
patients, the major cross-match test indicates compat- had recurrent fludarabine-associated AIHA and who
ibility, and transfused RBC can be expected to survive was challenged with the adenosine deaminase
normally. inhibitor pentostatin, a potent immunosuppressant
Laboratory and clinical features of methyldopa- with no known fludarabine cross-reactivity. Orchard
induced AIHA are summarized in Table 8-9. and associates244 reported on a patient who had an
episode of severe AIHA 19 weeks after the last of three
courses of fludarabine; he did not respond to cortico-
AIHA CAUSED BY DRUGS OTHER steroids, but his hemoglobin returned to normal after
a splenectomy. Three months later, at a time when his
THAN METHYLDOPA hemoglobin and platelet count were normal, he was
treated with chlorambucil, 10 mg/day. After nine
A number of drugs other than methyldopa have days of treatment, his hemoglobin was normal, but
caused AIHA, as indicated in Table 8-8. the next day he was admitted in a moribund state
with a hemoglobin of 5.0 g/dL and evidence of severe
Purine Analogues hemolysis. Despite treatment with steroids, his hemo-
globin fell further, and he died.
The most common drug by far to cause AIHA in There are two reports of Evans’s syndrome compli-
current practice is a purine analogue, fludarabine.222-246 cating fludarabine treatment for CLL. Shvidel and col-
A British group reported that 14 of 66 patients (21%) leagues245 described a patient who, after two cycles of
with chronic lymphocytic leukemia (CLL) developed fludarabine, developed AIHA and a drop in platelet
AIHA following fludarabine therapy.230,240 Another count from normal levels to 12,000/μL. He recovered
study from France found that 8 of 36 patients (22%) from Evans’s syndrome after treatment with cyclo-
developed AIHA.237 It is sometimes difficult to be sporine and prednisone. Sen and Kalaycio246 reported
confident that fludarabine caused the AIHA, as on a 64-year-old woman who had a white count of
10–20% of CLL patients are said to develop AIHA at 150,000/μL, a hemoglobin of 11 g/dL, and a platelet
some stage of their disease; 1.8–35% of CLL patients count of 250,000/μL. She was treated with two cycles of
have been reported to develop positive DATs.231 Data fludarabine, and 10 days later her DAT was positive,
strongly implicating fludarabine administration as a the white blood cell count was 14,000/μL, hemoglobin
cause of AIHA were reported by Diehl and was 5.5 g/dL, and platelet count was 5000/μL. The
Ketchum.242 These authors reviewed reports indicat- patient improved during treatment with prednisone
292 Immune Hemolytic Anemias

but ultimately required a splenectomy before entering from IgG-sensitized cells reacted strongly with all
into a durable remission of hemolysis with successful RBCs except Rhnull cells. None of the patients devel-
withdrawal of her corticosteroids. oped overt AIHA.
Orchard and coworkers244 summarized the associa- Joseph248 reported that the incidence of a positive
tion of AIHA and fludarabine as follows. AIHA is DAT was 9.5% among patients taking levodopa for
unlikely to occur in patients with de novo CLL who longer than 90 days.
are being treated for the first time with fludarabine. Gabor and Goldberg249 reported a patient who
Among patients who have been treated with multiple developed AIHA after 4 years of therapy with levo-
courses of alkylating agents, however, exposure to dopa. Six weeks after discontinuation of the drug, the
fludarabine triggers AIHA in about 20% of cases. The hematologic values had returned to normal, and the
hemolysis is usually severe, difficult to treat, and can DAT and IAT were negative. Five months following
prove fatal. Autoimmune thrombocytopenia might reinstitution of the drug, AIHA recurred and again
also be present. After treatment with fludarabine, resolved on discontinuation of the drug.
patients with AIHA are liable to relapse if they are re- Anti-e specificity of levodopa-induced autoantibo-
exposed to fludarabine, cladribine, or pentostatin. dies was reported by Lindstrom and coworkers250 and
Also, other forms of potent immunosuppressive by Bernstein,251 and in the case reported by Territo
chemotherapy could cause a recurrence of fludara- and associates,252 an eluate reacted with all RBCs
bine-induced AIHA, adding support to the hypothesis except Rhnull cells.
that effects on the cellular immune system are the
basis for the development of AIHA in this setting. Procainamide
AIHA has also been associated with 2-chlorodeo-
xyadenosine (cladribine). In one study, only two of Kleinman and colleagues256 studied the prevalence of a
114 CLL patients (1.8%) with no AIHA before receiv- positive DAT and IHA among 100 patients receiving
ing cladribine developed AIHA after five and six procainamide and among 100 age- and sex-matched
courses of cladribine, respectively.229 On the other controls. There was a significant increase in the fre-
hand, 23 patients had signs of AIHA before treatment quency of a positive DAT among patients receiving
with cladribine; the hemolytic anemia resolved com- procainamide compared with controls; three patients
pletely for six patients and resolved partially for eight had AIHA. The mean duration of procainamide admin-
patients.229 Chasty and coworkers231 reported that istration was 23.8 months for the patients with positive
four of 19 CLL patients (21%) treated with cladribine DATs and 21.3 months for those with negative tests.
developed AIHA, which was the same incidence Eluates were prepared from the RBCs of 18 patients
reported by the same group for fludarabine.226 with a positive DAT and gave positive results in 10
Hamblin and coworkers235 reported that one patient instances, including all three patients with hemolytic
with fludarbine-induced AIHA had severe hemolysis anemia. Seven of the 10 positive eluates were panag-
after receiving cladribine subsequently. They sug- glutinins, and three showed Rh specificity, demon-
gested that any purine analogue could lead to exacer- strated by a lack of reactivity with Rhnull cells. In
bated hemolysis, and others have shown that even addition, there are multiple individual case reports of
conventional chemotherapy (e.g., chlorambucil-based AIHA caused by procainamide.253-255
regimes) can exacerbate hemolysis. The presence of antinuclear factor (ANF) or clinical
Fludarabine is the 2-fluoro derivative of adenine lupus syndrome was determined in 18 patients with a
avalinoside. The added fluoro group enables fludara- positive DAT.253 The prevalence of ANF was 78%,
bine to resist inactivation by adenosine deaminase, an which is similar to the rate previously reported for
enzyme found in high levels in lymphocytes.241 The patients receiving procainamide. None of the three
drug enters the cells in the form of fluoro-ara-A metabo- patients with hemolytic anemia had clinical evidence
lite and is phosphorylated to a triphosphate that of drug-induced lupus, but two of the 15 without HA
accumulates in cells and inhibits DNA and RNA syn- did have it.253
thesis.241 It might also activate apoptosis. Fludarabine
seems to exert an immunosuppressive effect on the CD4 Nonsteroidal Anti-inflammatory Drugs
subset of lymphocytes, thus leading to an impairment
in the CD4/CD8 ratio, which could influence the induc- Immune hemolytic anemia has been reported follow-
tion or recurrence of RBC autoantibody. ing administration of nonsteroidal anti-inflammatory
drugs (NSAIDs), but in many instances the serologic
testing has been incomplete and the presence of RBC
Levodopa autoantibodies has not been documented well. In a
Henry and associates247 studied 80 patients with detailed review in 1986, Sanford-Driscoll and Knodel257
Parkinson’s disease who received levodopa therapy concluded that mefenamic acid caused HA by an
and found three patients who developed a positive autoimmune mechanism but that there was insufficient
DAT caused by IgG sensitization after eight to information concerning ibuprofen, sulindac, naproxen,
11 months of therapy, and an additional two patients tolmetin, and feprazone to assign specific mechanisms
with complement-coated RBCs. Eluates prepared of immune hemolysis.
Drug-Induced Immune Hemolytic Anemia 293

There have been multiple reports since that sergide, chlordiazepoxide, interleukin-2, rituximab,
time,258-273 but no convincing evidence that these and fenfluramine, although for these drugs the char-
drugs induce true autoantibodies. Many of the acterization of the responsible antibodies as RBC
reports indicate that when autoantibodies are pres- autoantibodies is less definite.
ent, they are present together with drug-dependent Cimetidine therapy has been reported to be associ-
antibodies. ated with AIHA in one patient and with a case of
In a number of instances, hemolysis associated with hemolytic anemia with a negative DAT.288,289 The
the administration of NSAIDs was part of a complex authors of both reports express caution in implicating
clinical picture involving multisystem organ failure the drug, however. This caution was emphasized by
with renal insufficiency, shock, and disseminated Petz and associates.10
intravascular coagulation.272,273

Nomifensine IMMUNE HEMOLYTIC ANEMIA


AND/OR POSITIVE DATs ASSOCIATED
Nomifensine is an antidepressant drug that was used WITH THE CEPHALOSPORINS
for about 8 years in Europe and for a much shorter
time in the United States, having been withdrawn
from sale worldwide in 1986 because of reports of side Cephalosporins differ from penicillins only in having
effects, which included numerous cases of immune a six-numbered dihydrothiazine ring instead of the
hemolytic anemia. A variety of serologic reactions five-nucleated thiazoldine ring, and in their side-
were reported, with most patients having drug- chain groups (see Figs. 8-2 and 8-15). Thus, it is not
dependent antibodies and a syndrome of acute surprising that the cephalosporins also have induced
intravascular hemolysis as is typical of drugs reacting positive DATs, IHA, thrombocytopenia, and neu-
by the “immune complex” mechanism. There are tropenia. Nevertheless, cephalosporins were used for
multiple reports, however, of patients with nomifen- more than 25 years before immune cell destruction by
sine-induced immune hemolysis whose serum con- the cephalosporins became a problem.
tained only RBC autoantibodies, some of which The cephalosporins have been grouped into “gener-
showed some Rh specificity.274-276 ations” on the basis of their spectrum of activity
against gram-negative organisms.290,291 First-generation
cephalosporins were first used in the United States in
Alpha-Interferon 1964. Second-generation cephalosporins started to be
The occurrence of autoimmune disease among patients used in 1976 (cefotetan was used from 1985), and
receiving alpha-interferon (α-IFN) therapy has been third-generation cephalosporins came into common
reported in several studies, and included among such use in 1981 (ceftriaxone was used from 1984 onward).
side effects is the development of AIHA.277-287 Table 8-10 lists the cephalosporins according to their
The incidence of AIHA caused by α-IFN appears to generations and their respective proprietary names. In
be quite low. In a retrospective series of 11,241 con- general, the pharmacologic properties of the cephalo-
secutive patients with chronic viral hepatitis treated sporins are similar. Most are administered by parental
with α-IFN, Fattovich and colleagues283 observed two injection, have a plasma half-life of approximately 1 to
cases of AIHA. Andriani and associates278 observed 2 hours, and are excreted unchanged from the body
nine patients over a ten-year period. Seven of the mainly via the urinary tract. Six of the cephalosporins
patients had been receiving α-IFN for a lymphoproli- (cephalexin, cefadroxil, cephradine, cefaclor, cefurox-
ferative disorder, and two patients had chronic mye- ime axetil, and cefixime) are used orally and are less
logenous leukemia. In all cases, the onset of hemolysis likely to cause immune cytopenia because of the rela-
was acute and severe and was associated with a posi- tively lower dose used compared with those used
tive DAT. In five patients, the serum antibody was intravenously.
characterized and found to be a panagglutinin (IgG in
four cases and IgA in one case). The interval between Cephalosporin-Induced Positive DATs
the start of α-IFN and AIHA varied from 1 day to
38 months. All patients recovered after discontinua- In 1967, Molthan and colleagues149 and Gralnick and
tion of the drug and treatment with prednisone. coworkers150 reported that 81% and 40%, respectively,
Fornaciari and coworkers284 reported a patient in of patients receiving cephalothin developed positive
whom AIHA had occurred within one month after ini- DATs. The patients were all quite ill and were receiving
tiation of α-INF; after reintroduction of α-IFN 1 year large doses of intravenous cephalothin (8–14 g/day
leater, AIHA recurred within the same interval. and 2–8 g/day, respectively). Later workers found a
much lower incidence of positive DATs. Petz181 found
that only 3% of 124 patients receiving an average dose
Other Drugs of 6.3 g/day for an average of 5.5 days developed a
Still other drugs may cause AIHA, for example, chlor- positive DAT. Spath and coworkers152 found that 4%
promazine, diphenylhydantoin, methoin, methy- of 97 patients receiving an average of 6.3 g/day for
294 Immune Hemolytic Anemias

TABLE 8-10. CEPHALOSPORINS

Generic Names USA Proprietary Names Year First Used in United States

1st Generation
cephalothin Keflin, Seffin 1964
cephalexin Biocef*, Keflex* 1971
cefazolin Ancef, Kefzol, Zolicef 1973
cephapirin Cefadyl 1974
cephradine Anspor*, Velosef 1975
cefadroxil Duricef*, Ultracef* 1981
2nd Generation
cefamandole Mandol 1976
cefoxitin Mefoxin 1976
cefaclor Ceclor* 1979
cefuroxime Kefurox, Zinacef 1983
cefuroxime axetil Ceftin* 1983
cefonicid Monocid 1984
cefotetan Cefotan 1985
cefmetazole Zefazone 1989
3rd Generation
cefotaxime Claforan 1981
cefoperazone Cefobid 1982
ceftizoxime Cefizox 1983
ceftriaxone Rocephin 1984
ceftazidime Fortaz, Tazicef, Tazidime 1985
cefixime Suprax* 1989
4th Generation
cefepime Maxipime 1996

*Oral only

5.5 days developed positive DATs. Ferrone and col- Spath and colleagues152 suggest that the mechanism
leagues292 reported that 12% of 42 patients receiving might occur in vitro but that it might not be the most
cephalothin developed positive DATs. York and associ- common mechanism involved in the development of
ates293 and Fass and coworkers294 reported that 2.2% positive DATs in vivo and probably has never been the
and 8% of 37 and 75 patients, respectively, who were mechanism causing cephalothin-induced hemolytic
receiving cephaloridine developed positive DATs. anemia. The most common mechanism involved in
Schwarz and associates,295 van Winzum,296 and cephalothin-induced positive DATs and immune
Meyers297 reported that approximately 2.4% of patients hemolytic anemia seems to be similar to that described
receiving cefoxitin, 4% of patients receiving cephalexin for penicillin. The patient’s RBCs become coated in
and ceftazidime, and 6.4% receiving cefotaxime, vivo with cephalothin, and if IgG cephalothin (or peni-
respectively, developed positive DATs. See the earlier cillin cross-reacting) antibodies are present, the RBCs
discussions in this chapter on the differences between become sensitized with IgG. Although the incidence of
the early and later findings. IgG sensitization is similar to that observed with
Molthan and coworkers149 and Gralnick and associ- patients receiving intravenous penicillin, the positive
ates150 suggested that positive DATs associated with DATs are usually weaker and the incidence of
cephalothin were due to nonimmunologic adsorption hemolytic anemia is much lower. As will be seen later,
of a cephalothin-protein complex onto RBCs, the the second- and third-generation cephalosporins seem
protein portion of which would be detected by AGS. to react in vitro and in vivo in a different way than the
Further studies extended the original observations and first-generation cephalosporins (e.g., cephalothin). The
led to the suggestion that cephalothin might alter the DATs associated with hemolytic anemia due to second-
RBC membrane so that it could adsorb many normal and third-generation cephalosporins are usually strong
plasma proteins (rather than cephalothin-protein com- positives due to IgG + C3, or less commonly to C3 with
plexes as originally suggested) nonimmunologi- no IgG sensitization.104 The mechanisms involved in
cally.51,140,155 Most of the studies supporting these the positive DATs are often a combination of the peni-
concepts were performed using cephalothin-treated cillin type plus the “immune complex” type (see the
RBCs incubated with plasma in vitro. The results of discussion later in this chapter).
Drug-Induced Immune Hemolytic Anemia 295

Cephalosporin-Induced Immune TABLE 8-11. DRUGS (N = 101) THOUGHT TO BE


Hemolytic Anemia CAUSING DIIHA INVESTIGATED AT AMERICAN RED
Until 1987, very few cases of IHA due to cephalo- CROSS BLOOD SERVICES, LOS ANGELES
sporins were reported. Only five well-documented (1983–2002)
cases due to first-generation cephalosporins were
reported in a 25-year period.183-186 All five of these cases Drug No. of Patients
were associated with cephalothin (one of these cases
cefotetan 68
was also associated with cefazolin).186 Several other ceftriaxone 10
cases (e.g., associated with cephalexin) have appeared tazobactam 4
in the literature, but the serologic evidence in these piperacillin 3
reports were incomplete.298,299 Second-generation clavulanate 3
fludarabine 2
cephalosporins began to be used in the late 1970s, but rifampicin 2
until 1987 there were only two case reports of a patient mefloquine 1
having IHA associated with their usage; these were due ticarcillin 1
to cefoxitin and cefamandole.187,300 The serologic evi- sulbacatam 1
dence in both these reports was not very convincing. cefoxitin 1
cefotaxime 1
For instance, in the cefamandole report, the DAT was erythromycin 1
only weakly positive with anti-IgG and stronger with probenecid 1
anti-C3; an eluate from the patient’s RBCs did not react procainamide 1
with cefamandole-treated RBCs; the serum reacted tolectin 1
with cefamandole-treated RBCs but only to a titer of 4,
and 1.5% of 344 random patients’ sera were found to
react with cefamandole-treated RBCs. All cases of IHA Second- and third-generation cephalosporins have
associated with cephalosporins reported up to this been by far the most common cause of DIIHA submit-
point appeared to be due to a mechanism similar to that ted to Dr. Garratty’s laboratory over the past 20 years
described for penicillin antibodies. The patients all had (Table 8-11). These 80 cases represent 80% of the 101
signs of extravascular RBC destruction associated with submitted DIIHAs. Of these, 68 cases (68%) were asso-
an IgG cephalosporin antibody on the RBCs and in the ciated with cefotetan, and 10 cases (10%) were associ-
serum. Four of the five patients had received penicillin ated with ceftriaxone.
prior to cephalosporin therapy; the antibody causing Most of the 69 cases reported in the literature (up to
the hemolysis, therefore, could have been a cepha- 2000) have been severe hemolytic anemias; 40% had
losporin antibody or a cross-reacting penicillin anti- fatal hemolysis. Table 8-12 lists the cephalosporins that
body. In 1982, Duran-Suarez and coworkers301 reported have been reported to cause IHA. This information
a positive DAT (IgG) following cefoxitin (a second-gen- could be misleading, as it is probable that only the most
eration cephalosporin) therapy; the patient had no dramatic cases have been reported. We suspect that
hemolytic anemia. The serum and an eluate from the many patients with a less severe HA due to these
patient’s RBCs reacted (titer of 1024) with allogeneic cephalosporins probably never get diagnosed. The
RBCs when cefoxitin was added but did not react with
cefoxitin-treated RBCs. Thus, this seems to be the first
report of a cephalosporin antibody reacting by the so- TABLE 8-12. CEPHALOSPORINS REPORTED
called immune complex mechanism. TO CAUSE IMMUNE HEMOLYTIC ANEMIA
The significance of the cephalosporins changed (UP TO 2000)
dramatically when, in 1987, Salama and colleagues302
Drug No. of Cases References
reported a case of severe intravascular hemolysis due
to a third-generation cephalosporin, cefotaxime. The cephalothin 5 183–186
DAT on the patient’s RBCs was due to RBC-bound cefazolin 1 186
complement; no RBC-bound IgG was detected. An cephalexin 1 294
eluate from the patient’s RBCs was nonreactive. The cefamandole 1 187
cefoxitin 2 98,300
patient’s serum reacted with allogeneic RBCs only in cefotaxime 3 86,330
the presence of cefotaxime; drug-coated RBCs did not ceftriaxone 18* 303,305,313–320,321,326,
react. These results were very different from those 327,332,334,337,339,341
seen with other cephalosporins and were unexpected cefotetan 31* 304–307,309–312,319,322,
323,328–331,333,334,
for this type of drug. Since this report, more than 341,34
50 additional cases of IHA associated with second- ceftizoxime 4 319,320,331
and third-generation cephalosporins have been cefixime 1 330
reported.86,87,98-102,302-345 Most of the cases were associ- ceftazidime 2 101,333
ated with severe hemolysis, and approximately 40% TOTAL 69
had fatal HA (associated with cefotetan, ceftriaxone, *Many other cases are reported, without individual case reports, in Arndt
and cefoxitin). et al.104 and Viraraghavan et al.343
296 Immune Hemolytic Anemias

same conclusion might apply to thrombocytopenia.


There are no published systematic studies to show the TABLE 8-14. PARAMETERS OF CEFOTETAN-
incidence of positive DATs or HA (possibly mild) after ASSOCIATED HEMOLYSIS IN 85 PATIENTS
therapy with commonly used cephalosporins such as WITH HEMOLYTIC ANEMIA*
cefotetan or ceftriaxone.
Viraraghavan and colleagues,343 from the US Food Parameter* Mean Range
and Drug Administration (FDA), reviewed 85 cases of
Decrease in hemoglobin levels
HA associated with cefotetan therapy (five of which (n = 20) 6.65 mg/dL 1.6–10.3 mg/dL
have been published) that had been reported to the Final hemoglobin levels
FDA’s Spontaneous Reporting System and the World (n = 52) 5.2 mg/dL 3–9 mg/dL
Health Organization’s database between December Increase in LDH levels
(n = 24) 188 U/L 0–8000 U/L
1985 and October 1997. Tables 8-13 and 8-14 show
some data from this study. The DAT was positive for LDH, lactate dehydrogenase; n, number of patients with value reported
50 of 52 patients tested, and cefotetan antibodies were * Other parameters included the result of a DAT (reported for 52 patients and
positive in 50 cases) and the need for transfusion (47 patients). Antibodies to
detected in 30 of 30 patients tested. New-onset renal cefotetan were reported in 30 patients and detected in 30 patients.
dysfunction was noted in seven patients (8.2%). There From Viraraghavan R, Chakravarty AG, Soreth J: Cefotetan-induced haemolytic
were 15 (18%) fatal HAs. Fifteen patients had received anaemia: A review of 85 cases. Adv Drug React Toxicol Rev
2002;21:101–107.
cefotetan previously, four of these had fatal hemolysis.
Tables 8-15 and 8-16 show some clinical and sero-
logic characteristics of the DIIHA associated with the
published cases of cefotetan and ceftriaxone. Just as coated RBCs (penicillin-type mechanism) but, unlike
there was a major difference between the clinical and penicillin or cephalothin, it also usually reacts with
laboratory findings associated with DIIHA caused by untreated RBCs in the presence of drug (“immune
first-, second-, or third-generation cephalosporins, complex” mechanism); in about one third of patients,
there are major differences between cefotetan (a drug-independent antibodies are also detected.104
second-generation cephalosporin) and ceftriaxone (a Also, unlike the case with penicillin or cephalothin,
third-generation cephalosporin). with cefotetan signs of in vivo intravascular lysis (e.g.,
Cefotetan appears to be extremely immunogenic in hemoglobinemia/hemoglobinuria) are also common.
that titers of cefotetan antibodies are sometimes as The clinical and laboratory findings associated with
high as 200,000, and DIIHA can occur after receiving a ceftriaxone-induced HA are very unusual. Children
single dose of cefotetan for the first time. The cefote-
tan antibody usually reacts best in vitro against drug-
TABLE 8-15. CLINICAL AND SEROLOGICAL
FINDINGS ASSOCIATED WITH CEFOTETAN-
TABLE 8-13. DEMOGRAPHIC CHARACTERISTICS INDUCED IHA
OF 85 PATIENTS WITH CEFOTETAN-ASSOCIATED
HEMOLYSIS* Approx. 80% of 31 patients received cefotetan for surgery; usually
a single dose of 2 g was used.
Characteristic A history of previous cefotetan therapy was not common.
HA was obvious in less than 1 day to 13 days after receiving
Agea (yr) cefotetan. Only two patients had HA in less than 1 day;
Mean 51.84 the mean of the other 29 was 9 days.
Range 21.91 Patients (n = 31) nadir Hb after receiving cefotetan = 2.6 g/dL
Sexb [n (%)] (mean = 4.8 g/dL).
Males 20 (24) Most patients had signs of intravascular lysis (hemoglobinemia/
Females 64 (76) hemoglobinuria).
Indication for cefotetanc [n (%)]
Fatal HA and renal failure occurred in 19% and 19% of patients,
Treatment 32 (41)
respectively.
Prophylaxis 47 (59)
Cesarean section 8 (10) Patients always have positive DAT:
Other obstetrics/gynecology 10 (13) 100% have RBC-bound IgG
Other surgery 29 (37) 86% have RBC-bound C3
Length of administrationd [n (%)] 44% have RBC-bound IgA
≤1 day 23 (30) 7.4% have RBC-bound IgM
>1 day 52 (70) All sera agglutinated cefotetan-treated RBCs (median titer 512) and
Prior cefotetan administration [n (%)] 15 (18) reacted by IAT (median titer 16,000); all normal sera also
Fatalities in patients with prior exposure [n (%)] 4 (27) reacted but were nonreactive when diluted ≥ 1:20.
All but one of the sera reacted with untreated RBCs in the presence
a n = 77 (age not stated for 8 patients)
b
of cefotetan (“immune complex” mechanism).
n = 84 (sex not stated for 1 patient)
c n = 79 (indication not stated for 6 patients) 33% and 40% of sera reacted with RBCs without drug being
d n = 75 (length of administration not stated for 10 patients) present, with saline-suspended RBCs or in the presence of
n = number of patients PEG, respectively.104
* From Viraraghavan R, Chakravarty AG, Soreth J: Cefotetan-induced
haemolytic anaemia: A review of 85 cases. Adv Drug React Toxicol Rev Clincal data from references 87, 98, 100, 103, 303–307, 309–312, 319,
2002;21:101–107. 322, 323, 328–331, 333, 334, 339. Serological data from reference 104.
Drug-Induced Immune Hemolytic Anemia 297

TABLE 8-16. CLINICAL AND SEROLOGICAL FINDINGS ASSOCIATED WITH CEFTRIAXONE-INDUCED IHA

Usually HA is more acute and severe in children313-317,321,327,335,339,341,344,345 [HA started 5 minutes to 7 days after receiving drug
(for 5 children a mean of 44 min, and 2 children a mean of 5 days)] compared with adults303,308,318,320,326,332,334,342 [HA started
30 minutes to 34 days (1 patient was 30 minutes, the mean for the other 7 was 12 days)].
Fatal HA occurred in 67% of 9 children and 38% of 9 adults.
History of previous ceftriaxone therapy.
Positive DATs associated with complement in all cases and IgG in 75%; none had detectable RBC-bound IgA or IgM.104
Serum and eluted antibodies only detected by immune complex method104 (serum + drug + RBCs). Three patients sera only reacted with
a metabolite of ceftriaxone (ex vivo antigen).321,332

often suffer dramatic acute hemolysis within a short To help interpret whether cross-reacting antibodies
time (e.g., 5 to 30 minutes) of receiving ceftriaxone; 70% are the cause of the positive reactions, it is useful to
of these are fatal HA. The patients usually have test penicillin-coated RBCs in parallel with the
received ceftriaxone therapy previously with no ill cephalosporin-coated RBCs. If positive results are
effects. Unlike cephalothin and cefotetan antibodies, obtained with the penicillin-coated RBCs, the only way
ceftriaxone antibodies are demonstrable in the labora- to prove specificity is to perform inhibition studies with
tory only by testing the patient’s serum in the presence the putative antibiotics or adsorb with antibiotic-coated
of drug (“immune complex” method). The sera do not RBCs. It is more difficult to differentiate nonspecific
react with ceftriaxone-treated RBCs (see Table 8-16). protein adsorption from reactions due to cephalosporin
Unfortunately, it has not been proven that ceftriaxone antibodies. Spath and colleagues152 used monospecific
will bond covalently to RBCs to provide in vitro ceftri- antiglobulin sera, such as antialbumin, to demonstrate
axone-coated RBCs, although one would expect this to that the AGT was positive due to nonspecific adsorp-
be the case. Bateman and associates345 found that 14% tion of normal plasma proteins rather than to RBC-
of 29 children with HIV infection made ceftriaxone bound anticephalothin.
antibodies, and another 24% were noninterpretable, as The nonimmunologic uptake of proteins onto RBCs
they had drug-independent auto- or alloantibodies can be prevented by diluting plasma/sera to a ratio of
present in their sera. 1:20 when working with cephalothin-treated RBCs,1,347
but a 1:100 dilution might be required when working
Problems with Interpreting Serologic with cefotetan-coated RBCs.342 Eluates (because of their
Results of Cephalosporin-Induced low protein content) do not show the effect. Thus, it can
Cytopenias generally be assumed that if sera diluted at ratios of
1:20 or 1:100, or eluates from AGT-positive RBCs, react
One must be quite careful in interpreting serologic with cephalosporin-coated RBCs, a cephalosporin (or
results involving cephalosporins in one’s own labora- cross-reacting penicillin) antibody is probably present.
tory or when reading published reports. As mentioned Another factor that could help considerably is the pH
earlier, RBCs that have been exposed to cephalothin used to prepare the cephalosporin-coated RBCs. Spath
can adsorb proteins nonspecifically from the and coworkers153 showed that penicillin-coated RBCs
plasma/serum, leading to the detection of such pro- that had been coated at pH 10.0 yielded penicillin anti-
teins by the AGT. Gralnick and associates,150 in their body titers 16 times that of titers using RBCs coated at
original 1967 report on cephalothin-induced positive pH 7.3. Cephalothin-coated RBCs prepared at pH 7.3
DATs, mentioned that 17 of 20 sera from healthy indi- and 10.0 showed similar titers with cephalothin anti-
viduals caused direct agglutination of cephalothin- bodies. As penicillin-coated RBCs are often tested in
coated RBCs and that all 20 reacted by the AGT. In 1971, parallel to cephalothin-coated RBCs, many workers, for
Gralnick and colleagues183 suggested that the aggluti- convenience’s sake, prepared RBCs coated with both
nation and AGT reactions with cephalothin-coated antibiotics at pH 8.5–10.0.1 Branch and coworkers154
RBCs were related to the level of IgM and IgG, respec- studied sera from 133 random patients and found that
tively, in the normal sera. Ferrone and coworkers346 also sera from 97% of these patients reacted, by AGT, when
correlated in vitro positive AGTs with cephalothin- tested against RBCs that had been coated with
coated RBCs with serum immunoglobulin concentra- cephalothin at pH 8.5. Only 4.6% of the sera reacted
tions. Abraham and colleagues142 tested 39 sera from against RBCs prepared with cephalothin at pH 6.0.
healthy hospital personnel and found that 69% reacted Thus, it would seem wise to prepare cephalothin- (and
with cephalothin-coated RBCs, but 77% of the same probably other cephalosporin-) coated RBCs at the
sera reacted with penicillin-coated RBCs. Thus, reac- lower pH; this should help considerably in alleviating
tions obtained with cephalothin-coated RBCs could be nonspecific uptake of proteins. One word of caution is
due to nonspecific adsorption of proteins, specific reac- that Branch and coworkers154 did not provide data to
tions due to cephalothin antibodies, or cross-reacting show that RBCs were as strongly coated with
penicillin antibodies. cephalothin at pH 6.0 as at pH 8.5; this is important to
298 Immune Hemolytic Anemias

maintain sensitivity for detecting cephalothin antibo- adding cephalosporin-treated RBCs in a hapten inhibi-
dies. We find a pH of 7.0–7.4 to be suitable for prepar- tion assay. Cefotetan, but not cephalothin, inhibited 18
ing cephalosporin-coated RBCs routinely. of 18 agglutinins and nine of 12 AGT reactions with
The relative efficiency of the cephalosporins (other cefotetan-treated RBCs. In contrast, although all the
than cephalothin) to cause nonspecific uptake of agglutination reactions with cephalothin-treated RBCs
RBC-bound proteins is not well documented in the could be inhibited by cephalothin, none of the AGT
literature. Kosaki and Miyakawa348 and Mine and reactions was inhibited. This suggests that some of the
coworkers349 showed that cephalexin and cefazolin, cefotetan-associated AGTs are due to antibody, but that
respectively, could cause nonspecific uptake of proteins all of the cephalothin AGTs are due to nonimmunologic
onto RBCs exposed to these cephalosporins in vitro. adsorption of protein, as suggested previously. Arndt
Branch and coworkers154 found that 20% of normal sera and Garratty351 suggested that the agglutination of
reacted with RBCs coated with cephapirin at pH 8.5, cephalosporin-treated RBCs was due to a “naturally
2% reacted with cephaloridine-coated RBCs, and none occurring” antibody that is present in a high percentage
reacted with RBCs coated with cefamandole, cepha- of the population (similar to the common occurrence of
lexin, or cefazolin at pH 8.5. Unfortunately, no controls penicillin antibodies). The presence of such antibodies
were included to show that the RBCs were indeed could be due to the presence of these drugs in our
coated with the latter cephalosporins. This is another natural environment (e.g., cephalosporins are added to
major problem with reports in the literature regarding cattle food).
reactions with antibiotic-coated RBCs. Most investiga- Arndt and Garratty351 suggested that the presence
tors incubate the putative antibiotic with the RBCs and of these “naturally occurring” antibodies might ex-
assume that the RBCs are coated; specific positive con- plain why such high titers (e.g., 200,000) of cefotetan
trols are often not available, but without such controls, antibodies and sometimes severe hemolytic anemia
negative results mean very little. occur after only a single dose of cefotetan.
Most of the reactions said to be due to nonspecific The foregoing discussion emphasizes that
adsorption of protein are AGT reactions, which can be cephalosporin-coated RBCs should always be tested
explained by the antiglobulin sera reacting with RBC- against as many control sera as possible from indi-
bound proteins that are not IgG cephalosporin anti- viduals not receiving the cephalosporin, and/or the
bodies. Neverthless, cephalosporin-coated RBCs often patient’s serum should be tested diluted at ratios of
are directly agglutinated by normal sera. Gralnick and 1:20 or 1:100 for cefotetan-coated RBCs, to avoid
coworkers150 reported that 17 of 20 normal sera (85%) nonspecific uptake of proteins or reactivity with
agglutinated cephalothin-coated RBCs but that only naturally occurring cefotetan antibodies. Clinicallly
one of these sera agglutinated penicillin-coated RBCs. significant antibodies will always exceed these dilu-
Garratty and associates350 tested 30 normal sera with tions. Such results will help put into perspective the
RBCs coated with cephalothin and cefotetan at pH 7.2 positive reactions obtained with serum from a patient
and pH 9.8 and found that almost all of the sera with cephalosporin-induced cytopenia.
directly agglutinated and yielded positive IATs when Arndt342 described three educational cases sent to
cephalothin- and cefotetan-treated RBCs were pre- Dr. Garratty’s laboratory for investigation of
pared at pH 9.8; and even at pH 7.2, most sera still cephalosporin-induced hemolytic anemia.
reacted. Far fewer reactions were seen with cefo-
taxime and ceftazidime-treated RBCs at either pH (see
Table 8-9). Combining these data with those of Branch P A T I E N T 1 : The patient, a 31-year-old female, deli-
and coworkers,154 it appears that cephalothin and vered her third child by cesarean section on March 4. She
cefotetan lead to a high percentage of false-positive was discharged from the hospital on March 7 with a hemo-
reactions; cephapirin to a moderately high percentage, globin of 8.9 g/dL but was readmitted on March 10 with
and cephaloridine, cefamandole, cephalexin, cefa- hemolytic anemia (hemoglobin 8.7 g/dL, reticulocytes
zolin, cefotaxime, and ceftazidime to a low percentage 10.5%, total bilirubin 3.3 mg/dL, LDH 503 U/L, hapto-
of false-positive reactions. It is unclear why globin <6 mg/dL) and a positive DAT (3+ with anti-IgG).
cephalosporin- (e.g., cephalothin- and cefotetan-) Her serum contained anti-C and anti-e that had been
treated RBCs should be agglutinated directly by identified previously, and she had a history of a previous
normal sera, even if they have adsorbed normal hemolytic anemia. On March 13, the hemoglobin dropped
plasma proteins from the sera. further to 6.1 g/dL, and the patient was transfused with
Arndt and Garratty351 reported that 53 of 72 (74%), 28 2 units of RBCs.
of 72 (39%), and 2 of 72 (3%) normal donor sera directly The hospital blood bank technologists suspected that
agglutinated cefotetan- (prepared at pH 7.3), this patient had an antibody to cefotetan, as she had
cephalothin- (prepared at pH 7.3), or penicillin- (pre- received two doses of that drug at the time of her surgery.
pared at pH 9.8) treated RBCs, respectively. Positive These blood bank technologists had seen a previous
AGTs were obtained with 63 of 72 (90%) and 72 of 72 patient with IHA due to cefotetan just 2 months previously,
(100%) normal donor sera incubated with cefotetan- or so they were aware of these types of cases. The blood
cephalothin-treated RBCs, respectively. Cefotetan and bank still had samples from the time of this patient’s
cephalothin were added to the reactive sera before surgery. The DAT was negative the day before the surgery,
Drug-Induced Immune Hemolytic Anemia 299

before she received her first dose of cefotetan. It was 2+ ration is not always helpful, the type of wash solution that
the day after the surgery, after she had received her is used can be important. In general, we have found 4°C
second dose. low ionic-strength saline (LISS) to be better than phosphate-
The hospital technologists reviewed the history of previ- buffered saline (PBS) as a wash solution when performing
ous hemolytic anemia and found that 3 years earlier, the acid eluates using commercial kits (we stopped using the
patient had had a cesarean section when delivering her commercial kit wash solution because of the problem of
second child and had received one dose of cefotetan at false-positive eluates in the presence of high-titer serum
that time. Thirteen days after that surgery, she was re- antibodies).353 When LISS was used as a wash solution
admitted with hemolytic anemia (hemoglobin 5 g/dL, in cases of cefotetan-induced IHA, less strongly reactive
reticulocytes 15%, haptoglobin <6 mg/dL) and received last washes were seen.352 This could be because low-
4 units of RBCs. In retrospect, the cause of this previous ionic solutions help retain more low-affinity antibodies on
hemolytic anemia could also have been an antibody to the RBCs during the washes. The patient’s March 10
cefotetan. serum reacted to a titer of 512 (AGT) vs. untreated RBCs
Further studies of this patient’s most recent sample in the presence of cefotetan; enzyme-treated RBCs were
showed the presence of IgG (4+), C3 (2+), and IgA (3+) strongly agglutinated; no hemolysis of test RBCs was
on her RBCs. Arndt and colleagues104 found RBCs from observed.
patients with cefotetan-induced IHA to be coated with IgG In conclusion, this patient had a high-titer anti-cefotetan
100% of the time, with C3 86% of the time, with IgA 44% antibody present in her serum, and her RBCs also were
of the time, and with IgM 7% of the time. coated with anticefotetan. This antibody most likely
The patient’s March 3 (presurgery, predrug) serum caused both the current hemolytic episode and the hemo-
sample was shown to contain anticefotetan, which lysis noted 3 years previously. The patient was warned
reacted with cefotetan-treated RBCs to a titer of 512 by never to receive cefotetan again, as that could lead to
AGT (untreated RBCs tested in parallel were nonreactive). more severe, possibly fatal hemolytic anemia.
The presence of preformed anticefotetan in this sample
thus confirmed that the hemolytic anemia seen 3 years Some important points about cefotetan-induced IHA:
previously was likely due to cefotetan. The patient’s
March 10 (postdrug) serum sample reacted much more • Cefotetan is a commonly used antibiotic; it is used
strongly; the anticefotetan hemolyzed, agglutinated, and prophylactically for surgeries (e.g., cesarean sec-
sensitized cefotetan-treated RBCs to titers of 128, 1024, tions), and the usage is not always known to the
and 128,000, respectively. The March 10 serum also attending physician after surgery.319
reacted weakly with untreated RBCs by AGT. The pres- • A single dose of cefotetan can result in dramatic
ence of an autoantibody (i.e., a drug-independent anti- hemolytic anemia. In some cases, the hemolytic
body) is not an uncommon finding in cases of IHA due to anemia lasts longer than expected.
cefotetan. Arndt and coworkers104 found autoantibodies • The hemolytic anemia usually becomes clinically
in about one third of the patients with cefotetan-induced apparent about one to two weeks after receiving
IHA that they have studied (and in up to 44% of patients, cefotetan. Unfortunately, patients sometimes receive
if PEG was used in the test system). more cefotetan at readmission (e.g., if an infection is
An acid eluate prepared from the patient’s RBCs suspected).
reacted with cefotetan-treated RBCs both when anti-IgG • The DAT is positive; this can range from strongly
and a specially standardized anti-IgA (see Chapter 6) positive (4+) to only weakly positive.104
were used, indicating that the anticefotetan had both IgG • Autoantibody (drug-independent antibody) can be
and IgA components. Anti-IgA is not used routinely to test present in the patient’s serum and/or eluate.104
eluates but was tested in this case to determine whether Thus, this drug-induced IHA can be confused with
the IgA coating the patient’s RBCs had anticefotetan warm AIHA, or if the patient was transfused (e.g.,
specificity. during surgery when cefotetan was given), a
When eluates are tested against cefotetan-treated delayed hemolytic transfusion reaction might be
RBCs, it is important always to test the last-wash super- suspected initially.
natant control in parallel. Many times, this last-wash • The serum antibody (anticeftotetan) typically reacts
control reacts with cefotetan-treated RBCs (Arndt and to a very high titer with (and might completely
coworkers104,352 found that 74% of eluates did so; 57% hemolyze) cefotetan-treated RBCs.104
of these reacted ≥1+). Although this reactivity probably • The serum antibody (anticefotetan) usually also
relates to the fact that serum cefotetan antibodies typically reacts by the “immune complex” method, but more
have very high titers (range, 4000–262,000; median = weakly.104
20,000), there is no direct relationship between the serum • Eluates prepared from the patient’s DAT-positive
anticefotetan titer and the presence of a reactive last RBCs react strongly with cefotetan-treated RBCs. In
wash.352 A relationship was seen between the presence a large percentage of cases, the last-wash eluate
of a reactive last wash and the strength of the patient’s control also reacts with cefotetan-treated RBCs,
DAT and/or the presence of autoantibody in the patient’s although usually weakly so. LISS appears to be a
serum or eluate.352 Although increasing the number of better wash solution than PBS for attempting to
times the patient’s RBCs are washed prior to eluate prepa- mitigate this problem.352
300 Immune Hemolytic Anemias

P A T I E N T 2 : The patient, a 76-year-old male, was cleared by day 5. If autoantibody had been present it
admitted with a diagnosis of pneumonia. On day 1, his would have still been detectable in the day 5 sample. If the
hemoglobin was 11.8 g/dL, creatinine 0.4 mg/dL, and anticeftriaxone had indeed been reacting with ceftriaxone-
serum and urine were clear. He was started on ceftriaxone treated RBCs, it should also have been detected in the day
(Rocephin; Hoffman-LaRoche, Nutley, NJ). On day 2, his 5 sample. Note that as no anticeftriaxone has ever been
creatinine was 1.0 mg/dL, and hemoglobinemia was shown to react with ceftriaxone-treated RBCs, no positive
noted. On day 3, his hemoglobin was 8.8 g/dL, hemat- control was available to prove that these treated RBCs were
ocrit was 23%, total bilirubin was 1.5 mg/dL, and hemo- indeed coated with ceftriaxone.
globinemia, hemoglobinuria, and oliguria were noted. In conclusion, this patient developed ceftriaxone
The ceftriaxone was stopped. antibody that caused intravascular hemolysis and renal
On day 4, his hematocrit was 21.2%, LDH was failure.
2858 U/L, serum hemoglobin was 92.9 mg/dL, hapto-
globin was 16 mg/dL, and creatinine was 4.3 mg/dL. Some important points about ceftriaxone-induced
He was transfused 2 units of RBCs. Over the next IHA:
4 days, plasmapheresis was performed three times. The
patient was discharged on day 11 with a creatinine of • The patients typically have received multiple doses
8.9 mg/dL and no recovery of his renal function. of ceftriaxone previously.
The patient’s DAT results were negative on day 1 (prior • In adults, the reaction tends to become apparent
to drug administration), 21⁄2+ with anti-C3 only on day 2, after the patient has received the drug for a day or
and then 1+ with anti-IgG and 3+ with anti-C3 on days two. In children, the reactions tend to be dra-
3 and 4. This patient’s hemolytic anemia was suspected matic, occurring within minutes of receiving
to be due to anticeftriaxone. He had a history of multiple ceftriaxone.313-317,327-339,344,345
previous ceftriaxone treatments. Typically, patients with • The DAT is positive due to C3 or IgG plus C3. The
IHA due to ceftriaxone have received multiple doses of the eluate is usually nonreactive.
drug. In some of the reported cases, patients have had • Ceftriaxone antibodies have been demonstrated
previously unrecognized hemolytic episodes. only by the “immune complex” method (enzyme-
All ceftriaxone antibodies reported so far have reacted treated RBCs react better than untreated RBCs);
only by the “immune complex” method (i.e., patient’s drug-treated RBCs are nonreactive. In two reported
serum plus drug plus RBCs); ceftriaxone-coated RBCs do cases, antibody was demonstrable only in the pre-
not react or ceftriaxone does not bind efficiently to RBCs. sence of “ex vivo” drug (urine from patients receiv-
When sera from patient 2 were tested by the “immune ing ceftriaxone).321,332
complex” method (i.e., in the presence of ceftriaxone • Reactivity of the patient’s serum against untreated
[1 mg/mL] against untreated RBCs), agglutination was RBCs without drug being present can be due to cir-
noted (titers = 4–16); the IAT was negative to only very culating drug-antibody immune complexes (if tran-
weakly positive. Controls of patient’s sera + PBS instead sient) or due to autoantibody (if persistent). (This is
of drug were nonreactive, therefore, this patient’s serum true of any drug, not just of ceftriaxone.)
contained anticeftriaxone.
Marked differences in reactivity can be observed when P A T I E N T 3 : The patient, a 59-year-old woman, had
testing enzyme-treated RBCs by the “immune complex” surgery about two weeks previously and then developed
method.104 For example, one patient’s anticeftriaxone a postoperative infection. On December 20, her hemo-
reacted to titers of 4/0 (agglutination/antiglobulin test) globin was 10.3 g/dL and she received 1 g of ceftria-
against untreated RBCs but reacted to titers of 256/1024 xone. On December 21, her hemoglobin had dropped to
against enzyme-treated RBCs.104 Unfortunately, titrations 8.2 g/dL and she received another 1 g of ceftriaxone.
of the sera from patient 2 against enzyme-treated RBCs On December 22, her hemoglobin had dropped further
were not performed. The agglutinin of patient 2 was inhi- to 5 g/dL and the ceftriaxone was discontinued. Her DAT
bited by treatment with 0.01M dithiothreitol (DTT), and was positive, reticulocyte count was 3.7%, and she was
therefore appeared to be an IgM antibody. And similar to transfused 4 units of RBCs.
other cases of IHA due to ceftriaxone, the eluate was non- When the hospital blood bank was called to verify the
reactive. patient’s identification (she had the same name as another
Patient’s sera were tested against RBCs that had been patient we had previously worked up with an IHA due to
treated with ceftriaxone in PBS or barbital buffer. Sera from anticefotetan), we were told that the doctor remembered
day 3 and day 4 agglutinated (2+ and 1+, respectively) that this patient had received ceftriaxone with her bowel
not only drug-treated but also untreated RBCs; sera from day surgery a few weeks earlier. Thus, this patient’s history
5 was nonreactive. As the patient received his last dose of was what we might expect with a ceftriaxone antibody—
ceftriaxone on day 3, it was suspected that the positive the patient had received the drug previously, and the reac-
results seen on days 3 and 4 were due to circulating drug- tion had taken a day or two to become apparent (as seen
antibody immune complexes that were still present in the in adults).
samples from those days (the half-life of ceftriaxone is about The patient’s DAT was positive (anti-IgG 1+, anti-C3
9 hours in elderly subjects and about 15 hours in patients 3+), but the “immune complex” testing in the presence of
with impaired renal function). These immune complexes had ceftriaxone was negative. Thus, this patient did not have
Drug-Induced Immune Hemolytic Anemia 301

a ceftriaxone antibody. It was suspected that the history penicillin has been reported, but the exact incidence is
might not have been accurate—that the patient had unclear.355,356 Cerny and Pichler357 believe that the
received a drug other than ceftriaxone (e.g., cefotetan) interpretation of available data is controversial, and
during the surgery a few weeks previously. The occur- although some believe it is safe to administer a
rence of hemolysis a couple of weeks after surgery is what cephalosporin to a patient with penicillin allergy, they
would be expected for a case of cefotetan-induced IHA. still recommend an immunologically unrelated anti-
The patient’s serum and eluate were tested against biotic for patients with penicillin or cephalosporin
cefotetan-treated RBCs and found to contain anticefotetan. allergy.
The undiluted serum hemolyzed cefotetan-treated RBCs and, There are some data in the literature relating to cross-
when diluted, reacted to titers of 320 and 10,240 (agglu- reactivity of penicillin and various cephalosporin
tination test and antiglobulin test, respectively). Untreated antibodies with respect to RBC interactions. Abraham
RBCs were nonreactive. The hospital blood bank was con- and coworkers358 described a severe cutaneous reaction
tacted and asked to determine whether the patient had to cephalothin in a patient who previously had re-
received cefotetan during the surgery. They checked the sur- ceived penicillin repeatedly. The patient had a positive
gical records and discovered that the patient had received skin test with cephalothin but not with penicillin. The
2 g of cefotetan for her surgery on December 10; she had patient also had an antibody that reacted with
been readmitted on December 20, exactly 10 days later. cephalothin but not with penicillin-treated RBCs. The
Luckily, she did not receive more cefotetan at the time of patient subsequently was given penicillin with no reac-
readmission. tion. Petz359 showed that penicillin and cephalothin
In conclusion, this patient had cefotetan antibodies, not antibodies usually cross-react with penicillin and
ceftriaxone antibodies. cephalothin-treated RBCs, but in 40% of sera, strikingly
dissimilar titers are present. When penicillin was
An important point is illustrated by this case: administered to patients with antibodies to both types
of treated RBCs, 55% showed an increased titer with
• A good history is important but can be difficult to penicillin-treated RBCs, and 23% demonstrated an
obtain. When drug-induced IHA is suspected, it is increased titer with cephalothin-treated RBCs. After
important to find out not only what drug(s) the administration of cephalothin, 25% showed increased
patient is currently taking but also what the patient titers against cephalothin-treated RBCs and 14%
received (e.g., in surgery or in preparation for against penicillin-treated RBCs. Spath and associates153
surgery) a few weeks previously. This information showed that sera from patients in whom positive DATs
is often “hidden” in the anesthesiologist’s notes developed following cephalothin administration
and can require some detective work to discover. reacted with penicillin- and cephalothin-treated RBCs
to a similar titer. Adsorption of three serum samples
with penicillin-treated RBCs caused an almost com-
Cross-Reactivity of Cefotetan plete loss of activity, a moderate loss of activity, and no
and Ceftriaxone Antibodies Associated significant decrease in titer, respectively, against
with Hemolytic Anemia, with Other cephalothin-treated RBCs.153
Arndt and Garratty360 tested seven sera previously
Cephalosporins, and Penicillin identified to contain cefotetan antibodies and one
Penicillins and cephalosporins share a basic β-lactam serum previously identified to contain ceftriaxone
structure (see Fig. 8-2). Antibodies can be directed to antibodies, against RBCs coated with (or in the pre-
this basic structure or to side chains, which could be sence of) nine other cephalosporins, penicillin, and
specific to a certain antibiotic. Most penicillin- 7-amino-cephalosporanic acid in the presence of
sensitive patients develop antibodies to the β-lactam RBCs. They also used hapten inhibition to indicate
nucleus; thus, extensive cross-reactions are observed cross-reactivity. Sera containing cefotetan antibodies
with semisynthetic penicillins that have different showed some cross-reactivity only with cephalothin
side chains. Some patients develop antibodies exclu- and cefoxitin (and to a much lesser extent with peni-
sively against side chains (e.g., a patient might be cillin and ceftazidime). The ceftriaxone antibody
allergic to amoxicillin but not to penicillin G). showed very weak cross-reactivity only with cefo-
Cross-reactivity of penicillins and cephalosporins taxime, cefamandole, and cefoperazone.360
has been more controversial. Although there is evi- Compared with the other cephalosporins, cefotetan
dence that antibody responses to penicillins and has a unique R1 group (Fig. 8-15). The portion(s) of the
cephalosporins are closely linked,354 the incidence of cefotetan molecule against which the patient’s anti-
reactions to cephalosporins in patients with a history bodies are directed is unknown. Both cephalothin and
of penicillin allergy is low.354-359 Novalbos and col- cefoxitin showed the strongest cross-reactivity with
leagues356 reported that the risk of a patient with cefotetan antibodies. Their R1 and R2 groups are very
penicillin allergy having an allergic reaction to similar to each other but are quite different from those
cephalosporins was very low if the cephalosporins in cefotetan. Cefamandole, which has the same R2
had a side chain different from the responsible peni- structure as cefotetan, was only weakly cross-reactive.
cillin. Cephalosporin allergy in patients not allergic to Cefamandole, cefoperazone, and cefotaxime showed
302 Immune Hemolytic Anemias

Cephem nucleus 7-ACA


1 H H
S S
R1 C NH7 H2N
N4 N O CH3
O
O R2 O
COO– COOH O

Cefotetan Ceftriaxone
O H3C N OH
N N
N C N
H2NC S N
N N
C C C CH2S H2N S N
OCH3 CH2S O
HOOC S CH2

R1 R2 R1 R2

Cephalothin Cefotaxime
O N C O
CH2OC N CH2OC
S H2N S
CH3 CH3 OCH3 CH3

R1 R2 R1 R2

Cefoxitin Cefoperazone
HO CH N N
O N
NHCO N
CH2OC CH2S
S D N
CH3 NH2 CH3

D N
R1 R2 R1 R2
C2H3

Ceftazidime Cefamandole
N N
N C CH2N CH N
N
N CH2S
H2N S OH
OC(CH3)2COOH CH3

R1 R2 R1 R2

Cefazolin Cefepime
N N N C
N H3C
N CH2 CH2S S N CH2N
CH3 H2N S
N N OCH3

R1 R2 R1 R2

Penicillin
O
H H
S
H CH3
N CH3
O
COOH
FIGURE 8-15. Chemical structures for 7-aminocephalosporanic acid (7-ACA), penicillin, the cephalosporin (e.g., cephem) nucleus, and the R1 and R2
side chains found in the cephalosporins that were tested in a cross reactivity study. (From Arndt PA, Garranty G: Cross-reactivity of cefotetan and
ceftriaxone antibodies, associated with hemolytic anemia, with other cephalosporins and penicillin. Am J Clin Pathol 2002;118:256–262.)
Drug-Induced Immune Hemolytic Anemia 303

weak cross-reactivity with ceftriaxone antibodies are no previous antibodies to use as controls, one
tested by mixing patient’s serum plus drug plus must test the patient’s serum against drug-treated
RBCs.360 Only cefotaxime and ceftriaxone share a RBCs and untreated RBCs, but if negative results
common R1 structure. are obtained, the results are of no value.
Thus, although it usually is recommended that 3. What concentration of drug should be used for
patients with cephalosporin-induced hemolytic “immune complex” testing? It is advisable to try
anemia never take any of the cephalosporins again, several concentrations.
there are few data to support that advice. The data of 4. If the clinical history is good and one cannot detect
Arndt and Garratty360 suggest that there is very little drug antibody, one should consider using metabo-
cross-reactivity with antibodies to the two most lites of the drug, either obtained from the drug
common cephalosporins (cefotetan and ceftriaxone) to manufacturer or as “ex vivo” metabolites from the
cause DIIHA and the other cephalosporins tested. urine and/or plasma of patients taking the drug.
Whether these in vitro data relate to in vivo reactivity
remains unknown. To interpret results accurately, one must have relevant
controls. Generally, the possible controls for detecting
drug-dependent antibodies are the following:
USEFUL SEROLOGIC METHODS
FOR INVESTIGATING DIIHA 1. The patient’s serum and/or eluate are tested
against untreated RBCs as a negative control when
Regardless of the controversies about the mechanisms drug-treated RBCs are used.
involved in drug immunology, serologic approaches 2. Drug-treated RBCs should also be tested against a
have remained very much the same. The basic sero- pool of normal inert sera as a negative control (if
logic techniques used to investigate DIIHA are given the patient’s serum is tested diluted 1:20 or 1:100,
in this section, but a few remarks regarding their use then the control normal sera pool should be appro-
are necessary as a preface (other specific remarks are priately diluted).
given in a “Notes” section following each technique). 3. The patient’s serum is tested in the presence of PBS
The first point to emphasize is that drug-induced as a negative control when the serum is tested in
IHA is rare. A positive DAT is much more likely to be the presence of drug.
due to causes other than drug-induced antibodies, 4. If possible, a positive control (i.e., an antibody
even when a patient is receiving any of the drugs against the appropriate drug) should be used; this
listed in Table 8-1. Even when an eluate prepared from is usually possible only in laboratories that special-
DAT-positive RBCs is nonreactive with reagent RBCs, ize in DIIHA, but cefotetan antibodies should be
the nonreactivity is much more likely to be due to the freely available from such laboratories.
eluate containing anti-A or -B from out-of-group
platelet or plasma transfusions than to a drug anti- One important point that is often not considered (even
body. One should also remember that up to 80% of all by purported experts in the field) is that it is optimal
positive DATs yield nonreactive eluates, and this is to use pure drug rather than the commercial product.
thought to be due to nonimmunologic uptake of IgG Commercially available drugs contain many inactive
onto patients’ RBCs when their plasma IgG levels are ingredients. In 1982, Brown364 reported that approxi-
high.361-363 If the eluate is reactive and the serum con- mately 800 such ingredients could be present in drugs
tains autoantibodies, it is much more likely that these available at that time. It is always possible that anti-
are associated with idiopathic AIHA than with drug- bodies demonstrated by the tests described in subse-
induced AIHA. quent pages of this chapter are directed against one of
We perform the time-consuming investigations for these ingredients rather than against the drug itself.
DIIHA only if the patient has definite evidence of a Law and associates365 reported on a patient who
hemolytic anemia and a good temporal relationship to developed HA during treatment with ibuprofen. The
treatment with a particular drug. If the putative drug patient’s serum and an eluate from the patient’s DAT+
has been reported as a cause for DIIHA, it is wise to RBCs reacted with RBCs only in the presence of the
follow the techniques used in the relevant report(s). If Motrin 400 (ibuprofen). Further studies showed that
the drug has never been reported, it is advisable to test the in vitro reactions were due to the orange dye
the patient’s serum and an eluate from the patient’s coating the Motrin tablets, not to the ibuprofen.365
RBCs against drug-treated RBCs and RBCs in the When the drug was stopped, the hemolysis resolved
presence of drug (“immune complex” technique). In and the serology became negative, suggesting that
such a case, the following questions or problems need this was indeed an IHA, but one that was due to an
to be addressed: antibody to the dye rather than to the drug.
Finally, if drug-independent antibodies are present,
1. Will the drug go into solution readily? it is sometimes necessary to adsorb them from the
2. Can drug-coated RBCs be prepared? What concen- patient’s serum using the same procedures discussed
trations of drugs and pH should be used? As there in Chapter 6 for removing autoantibodies. The serum
304 Immune Hemolytic Anemias

is then retested for the presence of drug-dependent PREPARATION OF DRUG-TREATED RBCs


antibodies.

Materials:
PREPARATION OF DRUG SOLUTIONS
1. Putative drugs.
2. Appropriate buffer, e.g.:
Materials: a. Phosphate-buffered saline (PBS), pH 7.3
b. 0.1M sodium barbital buffer
1. Putative drug(s), preferably in powder form. 2.06 g sodium barbital
Tablets and capsules can be used also. Least 100 mL deionized water
desirable is a drug that is already in solution or Adjust pH to 9.6–9.8 with 0.1 N HCl (appro-
suspension. ximately 1.5 mL)
2. Phosphate-buffered saline (PBS), pH 7.3, or other c. Potassium borate buffer
appropriate buffer as needed. 0.518 g boric acid
3. 0.1 N HC1; NaOH (to adjust pH if necessary). 0.22 g KCl
4. pH meter; 37°C incubator; scale, weigh boats/ 60 mL deionized water
paper, spatula. Adjust pH to 9.6–10.0 with 3 M NaOH
(approximately 2 mL)
Procedure: QS to 100 mL
3. Centrifuge; 37°C incubator (for some methods);
1. Check the solubility of the drug in the Merck refrigerator (for some methods); scale, weigh
Index366 or Physicians Desk Reference (PDR).367 boats/paper, spatula; pH meter and appropriate
2. a. If the drug is soluble in water, add the appro- reference buffer(s); test tubes (e.g., 13 × 100 mm)
priate amount of drug to the appropriate 4. Washed, packed group O RBCs, DAT-negative
amount of PBS (see the procedures for drug- (preferably fresh, e.g., less than 7 days old)
coated RBCs, “immune complex” method, or 5. Normal pooled serum/plasma
hapten inhibition). Mix well. 6. Alsevers or CPDA-1 solution
b. If the drug is somewhat soluble in water, add
the appropriate amount of drug to the appro- Procedure:
priate amount of PBS. Mix well. To attempt Refer to the previous section on preparation of drug
to increase solubility, incubate the solution solutions.
at 37°C and use a vortex mixer to mix
vigorously. Penicillin-Treated RBCs:
c. If the drug is not soluble in water, other sol-
vents might need to be tried (e.g., 1% albu- 1. Dissolve 120 mg penicillin G (or related drug,
min, alcohol, acetone, buffers of different e.g., ampicillin, methicillin, etc.) in 3 mL 0.1 M
pH).368-370 If solvents other than PBS are barbital buffer (40 mg/mL).
used, there could be a problem with isotoni- 2. Add 0.2 mL packed RBCs.
city. If so, the drug solution might need to be 3. In a separate tube, add 0.2 mL packed RBCs to
diluted in PBS or dialyzed against PBS (but 3 mL 0.1 M barbital buffer (control).
this could lead to precipitation of the drug). 4. Incubate at room temperature for 1 hour. Mix
d. If the drug is is in a suspension or solution, gently at regular intervals.
dilute it in PBS to achieve the desired 5. Wash four times with PBS.
concentration. 6. Suspend to 3%–5% with PBS.
3. If the drug does not go into solution completely,
centrifuge it and separate the clear supernatant Cephalosporin-Treated RBCs1:
into a new tube (when the drug does not com-
pletely go into solution, the supernatant might 1. Dissolve 100 mg cephalosporin in 2.5 mL pH
still contain enough drug for reaction with a 7.0–7.4 (e.g., 7.3) PBS (40 mg/mL).
drug antibody to be detected. Capsules and 2. Add 0.25 mL packed RBCs.
tablets contain fillers that might not go into solu- 3. In a separate tube, add 0.25 mL packed RBCs to
tion. This does not mean that the drug has not 2.5 mL PBS (control).
gone into solution). 4. Incubate at 37°C for 1 to 2 hours with occasional
4. If the pH of the drug solution is unknown, deter- gentle mixing.
mine the pH. If it is lower than 5 or higher than 5. Wash four times with PBS.
8 (using NaOH or 0.1 N HCl, respectively), 6. Suspend to 3%–5% with PBS.
adjust the pH to approximately 7.
Drug-Induced Immune Hemolytic Anemia 305

Nafcillin-Treated RBCs195,196: 2. Test normal pooled sera/plasma or PBS as a


negative control. For drugs that induce nonim-
1. Dissolve 68 mg nafcillin in 3 mL borate buffer. munologic protein adsorption (e.g. cefotetan,
2. Add 0.2 mL packed RBCs. cephalothin), PBS should be used as the negative
3. In a separate tube, add 0.2 mL packed RBCs to control.
3 mL borate buffer (control). 3. Repeat preparation of the drug-treated RBCs if
4. Incubate at room temperature for 1 hour. the controls do not react as expected.
5. Wash four times with PBS.
6. Suspend RBCs to 3%–5% with PBS. Notes:

Erythromycin-Treated RBCs371,372: 1. For some drugs (e.g., penicillin), a high pH


buffer is required for optimal coating of drug
1. Prepare 5 mL of a 2.5% solution of erythromycin onto RBCs. If a high pH solution is not available,
in PBS (see “Preparation of Drug Solutions”). PBS (pH 7–7.4) can be used (i.e., eluates from
2. If necessary, spin and remove supernatant from RBCs of a patient with a positive DAT due to
precipitate. penicillin antibodies react so strongly that they
3. Add 0.5 mL packed RBCs to 4 mL drug solution almost always react with penicillin-treated RBCs
supernatant. prepared at a less than optimal pH).
4. Add 0.5 mL packed RBCs to 4 mL PBS. 2. During preparation of drug-treated RBCs (e.g.,
5. Incubate at 4°C overnight. penicillin), if a small “clot” forms, remove it with
6. Wash four times using 4°C PBS. Remove RBCs applicator sticks before washing.
from drug precipitate (if present) after each wash. 3. Some drug solutions can cause variable hemoly-
7. Suspend RBCs to 3%–5% with PBS. sis of RBCs. Many procedures try to compensate
for this. For example, use of a lower incubation
RBCs Treated with Other Drugs: temperature (room temperature or 4°C) can help
alleviate some hemolysis.
1. If previous reports are available, check the liter- 4. If the patient to be tested has known alloanti-
ature with respect to the method used. bodies, select RBCs that do not possess the
2. If no previous reports are available, try treating appropriate antigens.
RBCs by one of the foregoing methods (see pro- 5. Cephalosporins (e.g., cephalothin [Keflin],
cedure for “Preparation of Drug Solutions”), cefotetan [Cefotan]) do not require a high pH for
and/or try the chemical cross-linking methods optimal coating. A lower pH (i.e., pH 6.0–7.4)
(see following) to bind the drug to the RBCs. decreases nonspecific protein adsorption seen
3. Variations of the foregoing procedures (e.g., when a high-pH buffer is used. The least amount
use of whole blood instead of packed RBCs) of nonspecific protein adsorption by drug-
can be tried, or a new method might need to be treated RBCs occurs if a pH 6 buffer is used, but
developed. this leads to a slight decrease in coating by drug,
thus we usually use pH 7.0–7.4 (Arndt and
Storage of Treated RBCs: Garratty, unpublished observations).
6. Do not prepare the drug solution until you are
1. For short-term storage (i.e., up to a week), pack ready to add RBCs. Some data (Leger and
RBCs (drug-treated and untreated) and add Garratty, unpublished observations) using
Alsevers or CPDA-1 solution. Store at 4°C. cefotetan showed that use of stored drug solu-
2. For long-term storage, pack RBCs (e.g., 1 mL) tion (e.g., five hours) resulted in significantly
and add Alsevers solution. Store frozen in liquid weaker drug-coated RBCs.
N2 or glycerol. 7. Although drug-treated RBCs can be stored short
term and long term, there could be weakening of
Quality Control: drug coating upon storage. Use of a positive
control (especially if a titration is performed) can
1. If a positive control (sera or eluate with previ- be useful in detecting such weakening of stored,
ously identified drug antibody) is available, test drug-treated RBCs.
against the drug-treated and untreated RBCs (see
the procedure for testing drug-treated RBCs).
306 Immune Hemolytic Anemias

TESTING OF DRUG-TREATED RBCs 5. A negative result of the patient’s serum/eluate,


in the presence of a positive result with the
positive control, indicates the absence of drug
Procedure: antibody.
6. Negative results of the patient’s serum/eluate in
1. If the drug (e.g., cefotetan) involved is known to the presence of a nonreactive positive control is
cause nonimmunologic adsorption of proteins, an invalid test. There might be no drug bound to
dilute the patient’s sera and the normal those RBCs.
sera/plasma pool (if tested) at a ratio of 1:20 (for 7. Negative results of the patient’s serum/eluate
cefotetan, we recommend a 1:100 dilution). The without a positive control (not available or no
positive control serum should also be diluted at previous case report) can only be interpreted to
a ratio greater than or equal to 1:20 or 1:100 in mean that antibodies to that drug were not
PBS. detected. Drug might or might not be bound to
2. For each sample to be tested (e.g., patient’s the test RBCs. Include a statement on the report
serum, patient’s eluate, patient’s last wash, to reflect that a positive control was not tested.
positive control, negative control(s) [i.e., PBS
and/or normal sera pool]), label two 10 × 75-mm Notes:
tubes: Label one set of tubes as drug-treated
(e.g., “DT”) and the other set of tubes as 1. This is a screening test for drug antibody. If the
untreated (e.g., “UT”). serum is shown to contain drug antibody, a titra-
3. Add two drops of serum, eluate, and so on to tion of the serum vs. drug-treated RBCs can be
each appropriate pair of labeled tubes. performed to determine the antibody’s titer. For
4. Add one drop of the appropriate 3%–5% RBC agglutination and IAT results, dilute the
suspension to each tube (i.e., drug-treated RBCs patient’s serum in PBS; for hemolysin results,
to the “DT” tubes and untreated RBCs to the dilute patient’s serum in fresh normal serum
“UT” tubes). (complement source) and read only for lysis.
5. Incubate at 37°C for one hour. 2. If plasma (e.g., EDTA-plasma) is used instead of
6. Centrifuge. serum, hemolysis of test RBCs due to drug anti-
7. Inspect all tests for hemolysis and/or agglutina- body will not be detectable.
tion. Record results. 3. If untreated RBCs are reactive with the patient’s
8. Wash RBCs four times with PBS. sample(s), determine whether alloantibody
9. Add antiglobulin sera. and/or autoantibody are present. If allo-
10. Centrifuge. antibody(ies) is (are) present, prepare new drug-
11. Read all tests for agglutination. All macroscopi- treated RBCs selecting those that are negative
cally negative results should be read microsco- for the appropriate antigens. If autoantibody
pically. Record results. is present, perform an adsorption(s) and then
12. Add antiglobulin control cells to all negative test the adsorbed sample. Alternatively, a dilu-
tests. tion of the patient’s sample could be selected, at
13. Centrifuge. which the alloantibody(ies) and/or autoanti-
14. Read for macroscopic agglutination. body no longer reacts, for testing against drug-
treated RBCs. A weak drug antibody could be
Interpretation: missed by this dilution method.
4. If the normal sera/plasma pool reacts with the
drug-treated RBCs but not the untreated RBCs, it
1. Hemolysis, agglutination, and/or a positive IAT
could be due to nonspecific protein adsorption
are considered positive results.
onto the drug-treated RBCs and/or to specific
2. A positive result of the patient’s serum/eluate
reactivity of one or more sera in the pool against
with drug-treated but not untreated RBCs indi-
the drug. One can test individual normal sera
cates the presence of drug antibody.
against the drug-treated RBCs. If only some sera
3. A positive result of the patient’s serum/eluate
react, then the second of the two explanations is
with both drug-treated and untreated RBCs
the most likely. If all sera react, then the first
indicates the presence of alloantibody and/or
explanation might be more likely. To overcome
autoantibody (see Note 3).
nonspecific protein adsorption, retest all sera
4. A positive result of the patient’s serum and the
(patient’s and control) at a 1:20 or 1:100 dilution
normal sera/plasma pool with drug-treated
(protein content should then be too low to be a
RBCs indicates the possible presence of non-
problem).
specific protein adsorption (see Note 4).
Drug-Induced Immune Hemolytic Anemia 307

USE OF CARBODIIMIDE TO COUPLE DETECTION OF ANTIBODIES TO DRUGS


DRUGS TO RBCs198,373 BY THE IMMUNE COMPLEX METHOD

Purpose of Test: Materials:


Carbodiimide is used to couple proteins and other
compounds (e.g., drugs) with a variety of functional 1. Drug, 1 mg/mL in PBS (see “Preparation of Drug
groups (i.e., carboxylic acids, amines, alcohols) to Solutions” procedure).
RBCs when the usual pH coupling method does not 2. Phosphate-buffered saline (PBS), pH 7.3.
work. 3. Fresh normal serum (source of complement [C]),
thawed.
Materials: 4. Pooled untreated and enzyme-treated group O
RBCs (e.g., antigen detection RBCs), antigen
1. Carbodiimide: 1-ethyl-3-(3dimethyl-aminopropyl) negative if appropriate.
carbodiimide HCl (stored frozen). 5. Polyspecific antihuman globulin serum; antiglo-
2. Drug to be coupled to RBCs. bulin control cells.
3. Group O reagent RBCs, washed and resuspended 6. 10 × 75-mm test tubes; 37°C incubator; serologic
to 50%. centrifuge; cell washer (optional).
4. Phosphate-buffered saline (PBS), pH 7.3.
5. 2% EDTA-PBS:
2 g EDTA, tetrasodium salt in 100 mL PBS Procedure:
Adjust the pH to 7.2 with 1 N HCl
Prepare this reagent ahead of time because it takes 1. Label two sets of 10 × 75-mm tubes for each drug
a while for the EDTA to go into solution; the to be tested: serum plus drug, serum plus PBS,
reagent is to be used cold (4°C). serum plus C plus drug, serum plus C plus PBS,
C plus drug, C plus PBS. Label 1 set of tubes
Procedure: “UT” (for untreated RBCs) and the other set of
tubes “ET” (for enzyme-treated RBCs).
2. To each tube add two drops patient’s serum,
1. Dissolve 20 mg of drug in 3 mL PBS.
two drops fresh normal serum (“complement”),
2. To a second tube, add 3 mL of PBS.
two drops drug solution, or two drops PBS as
3. Add 0.1 mL of 50% washed RBCs to each tube.
appropriate.
4. Freshly prepare the carbodiimide solution: Dissolve
3. To the set labeled “UT,” add one drop 6%–10%
50 mg of carbodiimide in 1 mL deionized H2O.
untreated RBCs. To the set labeled “ET,” add one
5. Add 0.5 mL carbodiimide solution to each tube.
drop 6%–10% enzyme-treated RBCs.
6. Incubate at 4°C for 50 minutes.
4. Incubate both sets of tubes at 37°C for 1 to
7. Pour the carbodiimide mixtures into a 16 ×
2 hours with gentle periodic mixing.
100 mm tube containing 5 mL of cold (4°C)
5. Centrifuge and examine tests for hemolysis and
EDTA-PBS.
agglutination. Record results.
8. Wash the RBCs three times in PBS.
6. Wash RBCs four times with PBS.
9. Test the drug-carbodiimide–treated RBCs, the car-
7. Add antiglobulin sera. Centrifuge and inspect
bodiimide-treated RBCs, and untreated RBCs
for agglutination (examine all macroscopically
using the desired method.
negative tests microscopically). Record results.
8. Add antiglobulin control cells to all nonreactive
Notes: tests. Centrifuge and inspect for agglutination.
Record macroscopically positive results with a
1. RBCs treated with carbodiimide without pro- checkmark.
tein tend to stick to glass tubes, similar to
glutaraldehyde-treated RBCs. Quality Control:
2. Use carbodiimide-treated RBCs as soon as pos-
sible after treatment. We have found that reactiv- 1. The tubes with serum plus C plus PBS should
ity can weaken within hours of treatment. normally be nonreactive. Reactivity in these
3. When coupling a cephalosporin (or other drug control tubes can be compared with the appro-
known to cause nonspecific adsorption of protein) priate test with drug solution. See interpretation.
to RBCs using the carbodiimide method, include 2. The tubes with C plus drug and C plus PBS
normal plasma/serum as a negative control for should be nonreactive. If positive and the
the test system. patient’s serum plus drug/PBS (without C in the
test system) was noninformative, repeat the test
with a new source of complement.
308 Immune Hemolytic Anemias

Interpretation: those dilutions by the foregoing procedure. The


Hemolysis, agglutination, or positive IATs might titer is the reciprocal of the last dilution that
occur together or separately. A positive result for any reacts 1+.
of the patient’s tests to which the drug was added, 3. If negative test results are obtained but the
and a negative (or significantly weaker) result for the patient’s clinical history warrants further evalu-
corresponding negative control tests (i.e., tubes in ation, the following should be considered:
which PBS was substituted for drug and tubes with a. Use of drug metabolites (ex vivo antigen),
no patient’s serum added) suggests the presence of such as urine or serum from (an)other
an antibody to the drug being studied. patient(s) taking the drug, instead of the
Examples of reactivity that might be seen and the drug solution; check previous publications
interpretation are shown in Table 8-17: for optimal source, timing of sample post
drug ingestion, etc. Urine might need some
Notes: processing (centrifuge to remove any precip-
itate/solid material, and check pH [see
1. The decision regarding whether an eluate from “Preparation of Drug Solutions” procedure,
the patient’s RBCs should be tested by this Note 4]).
method needs to be made on a case-by-case b. Use of different concentrations of drug (e.g., a
basis. saturated solution and dilutions of the drug).
2. If positive test results are obtained, the antibody c. Wash for the antiglobulin test using a solution
titer can be determined by preparing dilutions of of drug (e.g., 10 mg drug in 500 mL of PBS)
the patient’s serum in PBS (for agglutination and instead of PBS.
indirect antiglobulin test results) or in fresh
normal serum (for hemolysis results) and testing

TABLE 8-17. INTERPRETATION OF RESULTS OF IMMUNE COMPLEX METHOD

Example Serum ± C 37°C IAT Interpretation

1 + drug 0 0 Negative (no drug antibody present)


+ PBS 0 0
2 + drug 0 2+ Positive (drug antibody present)
+ PBS 0 0
3 + drug hemolysis +/– agglutination 1+ Positive (drug antibody present); weak auto- and/or alloantibody
also present
+ PBS 0 1+
4 + drug 0 1+ Negative (no drug antibody detected); weak auto- and/or
alloantibody present (could miss weak drug antibody)
+ PBS 0 1+
5 + drug 0 3+ Not interpretable due to presence of auto- and/or alloantibodies;
+ PBS 0 3+ test serum dilutions, adsorbed serum, and/or antigen-
negative RBCs
Drug-Induced Immune Hemolytic Anemia 309

DETECTION OF ANTIBODIES TO CEFOTETAN 4. Antiglobulin control cells should be macro-


scopically reactive.
5. Repeat the test if controls are not reacting as
Procedure: expected.

Interpretation:
1. Prepare (see separate procedure) or thaw cefote-
tan-treated and untreated RBCs (at least eight to 1. A positive reaction of the patient’s serum (i.e.,
ten drops of a 3%–5% v/v suspension of each undiluted serum = hemolysis, and/or the 1 in 100
will be needed). dilution of serum/plasma = agglutination and/or
2. Prepare a 1 in 100 dilution of patient’s serum/ positive antiglobulin test) or eluate with cefotetan-
plasma in PBS (e.g., 0.01 mL serum/plasma plus treated RBCs but not with untreated RBCs usually
0.99 mL PBS, or one drop serum/plasma plus indicates the presence of an antibody to cefotetan
nine drops PBS, then one drop of the 1:10 dilu- (see Notes 1, 2). Reactions with the untreated RBCs
tion plus nine drops PBS). could indicate the presence of alloantibod(y)(ies) or
3. For each sample to be tested, label two 10 × autoantibod(y)(ies) (see Notes 3, 4).
75-mm test tubes: cefotetan-treated (CT) or 2. No reaction of the patient’s serum and eluate with
untreated (UT). See sample worksheet. cefotetan-treated RBCs indicates that no cefotetan
4. Add two drops of patient’s serum, patient’s antibody is present.
diluted serum, diluted positive control, PBS,
eluate, or last wash to each of the two labeled Notes:
tubes.
5. To the set of tubes labeled “CT,” add one drop of 1. Sera/plasmas are diluted at least 1:100 to avoid
3%–5% (v/v) cefotetan-treated RBCs. the false-positive results (indirect antiglobulin
6. To the set of tubes labeled “UT,” add one drop of test) seen when testing undiluted serum/plasma
3%–5% (v/v) untreated RBCs. against cefotetan-treated RBCs. These false-
7. Incubate 60 minutes at 37°C. positive tests are due to direct agglutination or
8. Centrifuge and examine tests for agglutination nonimmunologic protein adsorption by the
and/or hemolysis. Record results. cefotetan-coated RBCs. There is usually not
9. Wash four times with PBS. enough protein in serum diluted 1 in 100 or in an
10. Add AHG or anti-IgG to all tubes. Centrifuge and eluate to cause this problem.
read for agglutination (check all macroscopically 2. The last-wash control can sometimes react with
negative results microscopically). Record results. cefotetan-treated RBCs. If this reactivity is weaker
11. Add AHG control cells to all nonreactive tests. than that of the eluate (i.e., eluate plus cefotetan-
Centrifuge and read. Record positive results treated RBCs = 2+ to 3+, last wash plus cefote-
with a checkmark. tantreated RBCs = w+), then the eluate can be
interpreted as positive. If the reactivity of the
Quality Control: eluate and last wash are similar, one can dilute
both the eluate and last wash (e.g., 1 in 10 or 1 in
20 in PBS) and repeat the test; if the diluted eluate
1. The positive control should be reactive with the
is reactive and the diluted last wash is non-
cefotetan-treated RBCs and nonreactive with the
reactive, then the eluate can be interpreted as pos-
untreated RBCs. The PBS-negative control
itive. Fewer strongly positive (>1+) last washes
should be nonreactive with the cefotetan-treated
were seen when RBCs were washed with LISS (vs.
and untreated RBCs.
PBS) when the Gamma Elu-Kit II was used for
2. The positive plasma control, when diluted 1 in
eluate preparation.352
100, should be strongly (3+ to 4+) reactive with the
3. If the patient has alloantibod(y)(ies) or autoanti-
cefotetan-treated RBCs but nonreactive with the
bod(y)(ies) that react(s) with untreated RBCs even
untreated RBCs. This expected reactivity provides
at the 1 in 100 dilution, prepare dilutions of the
a check for the stability of cefotetan-treated RBCs
patient’s serum (e.g., 1 in 500, 1 in 1000) and retest
that have been stored frozen and the reactivity of
against untreated and cefotetan-treated RBCs.
antibody that might have been frozen-thawed
Antibodies to cefotetan typically have titers in the
multiple times. If the reactivity of the plasma is
thousands; alloantibodies and autoantibodies
less than 3+ with cefotetan-treated RBCs (espe-
should not be reactive to such high titers.104
cially those that have been stored frozen), repeat
4. About 30% of patients with antibodies to cefote-
the test with a fresh aliquot of control serum/
tan also have drug-independent antibody that
plasma or with freshly treated RBCs.
reacts with untreated RBCs.104
3. A different positive control or a higher dilution
5. Antibodies to cefotetan usually also react by the
of the plasma might not react as strongly with
“immune complex” method.104
the cefotetan-treated RBCs.
310 Immune Hemolytic Anemias

2. If the serum plus drug reacts to a lower titer than


DRUG (HAPTEN) INHIBITION193
the serum plus PBS (a difference of at least two
dilutions), then inhibition by the drug has
occurred.
Materials:
3. If the serum plus drug reacts to a higher titer
than the serum plus PBS, then the drug antibody
1. Drug solution, 10 mg/mL in PBS (see “Prepara-
works preferentially by the “immune complex”
tion of Drug Solutions” method).
method.
2. Phosphate-buffered saline (PBS), pH 7.3.
3. Drug-treated RBCs (see method for “Preparation
Notes:
of Drug-Treated RBCs”), 3%–5% in PBS.
4. 37°C water bath/incubator; cell washer (optio-
1. IgG antibody is more difficult to “neutralize”
nal); serologic centrifuge.
than IgM antibody of the same titer.145 A
5. Antihuman globulin, polyspecific or anti-IgG;
checkerboard approach can be used—dilutions
antiglobulin control cells.
of the patient’s serum and different concentra-
tions of the drug. Alternatively, a dilution of the
Procedure:
patient’s serum that reacts 2+ could be selected,
and dilutions of drug could be added to that.
1. Prepare a master serial dilution of the patient’s
serum in PBS, at least one tube past the titer end-
point. Prepare enough of each dilution for a PBS
control and one drug test (e.g., 0.3 mL serum
R E F E R E N C E S
plus 0.3 mL PBS). 1. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias,
2. For each dilution to be tested, label two tubes 1st ed. New York: Churchill Livingstone, 1980.
(e.g., with dilution and “test” or “control”). 2. Pirofsky B: Autoimmization and the autoimmune hemolytic
3. Add 0.1 mL of each serum dilution to the two anemias. Baltimore: Williams & Wilkins, 1969.
3. Snapper I, Marks D, Schwartz L, et al: Hemolytic anemia
appropriate labeled tubes. secondary to mesantoin. Ann Intern Med 1953;39:619.
4. To the tubes labeled “test,” add 0.1 mL of drug 4. Harris JW: Studies on the mechanism of a drug-induced
solution. Mix. hemolytic anemia. J Lab Clin Med 1956;47:760.
5. To the tubes labeled “control,” add 0.1 mL of 5. Dausset J, Contu L: Drug-induced haemolysis. Ann Rev Med
PBS. Mix. 1967;18:55–70.
6. Worlledge SM: Immune drug-induced haemolytic anaemias.
6. Incubate 1 hour at 37°C. Sem Hematol 1969;6:181–200.
7. Add one drop of 3%–5% drug-treated RBCs to 7. Garratty G: Current viewpoints on mechanisms causing drug-
all tubes. Mix. induced immune hemolytic anemia and/or positive direct
8. Incubate 1 hour at 37°C. antiglobulin tests. Immunohematology 1989;5:97.
8. George JN, Raskob GE, Shah SR, et al: Drug-induced throm-
9. Centrifuge and examine for agglutination. bocytopenia: A systematic review of published case reports.
Record results. (This step is optional if the anti- Ann Intern Med 1998;129:886–890.
body is not an agglutinin.) 9. Breckenridge A, Dollery CT, Worlledge SM, et al: Positive
10. Wash RBCs four times with PBS. direct coombs tests and antinuclear factor in patients treated
11. Add two drops antihuman globulin serum. with methyldopa. Lancet 1967;16:1265–1268.
10. Petz LD, Gitlin N, Grant K, et al: Cimetidine-induced
12. Centrifuge and examine for agglutination. hemolytic anemia: The fallacy of clinical associations. J Clin
13. Add one drop antiglobulin control cells to all Gastroenterol 1983;5:405–409.
negative tests. 11. Landsteiner K: The Specificity of Serological Reactions (revised
14. Centrifuge and read for macroscopic agglutina- ed). Cambridge, MA: Harvard University Press, 1947:85.
12. Eisen HN, Carsten ME, Belman S: Studies of hypersensitivity
tion. Record positive results with a checkmark. to low molecular weight substances. III. The 2,4-dinitrophenyl
group as a determinant in the precipitin reaction. J Immunol
Quality Control: 1954;73:296.
13. Park BK, Coleman JW, Kitteringham NR: Drug disposition and
1. Dilutions of the patient’s serum plus PBS should drug hypersensitivity. Biochem Pharmacol 1987;36:581–590.
14. Petz LD, Branch DR: Drug-induced immune hemolytic
react with the drug-treated RBCs to a titer anemia. In Chaplin H Jr (ed): Immune hemolytic anemias.
similar to the previously determined titer. New York: Churchill Livingstone, 1985:47–94.
2. The antiglobulin control cells should be macro- 15. De Weck AL: Low molecular weight antigens. In Sela M (ed):
scopically reactive. The Antigens, vol II. New York: Academic Press, 1974:142.
16. Plescia OJ, Palczuk NC, Braun W, et al: Antibodies to DNA and
3. Repeat test if any controls do not react as a synthetic polydeoxyribonucleotide produced by oligodexyri-
expected. bonucleotides. Science 1965;148:1102.
17. Plescia OJ, Palczuk NC, Cora-Figueroa E: Production of anti-
Interpretation: bodies to soluble RNA (sRNA). Proc Natl Acad Sci USA
1965;54:1281–1285.
18. Kitteringham NR, Christie G, Coleman JW, et al: Drug-protein
1. If the serum plus drug reacts to the same titer as conjugates—XII. A study of the disposition, irreversible
the serum plus PBS, then no inhibition by that binding and immunogenicity of penicillin in the rat. Biochem
drug has occurred. Pharmacol 1987;36:601–608.
Drug-Induced Immune Hemolytic Anemia 311

19. Kristofferson A, Ahlstedt S, Svard PO: Antigens in penicillin 44. Miescher PA, Pepper JJ: Drug-induced immunologic blood
allergy. II. The influence of the number of penicilloyl residues dyscrasias. In Miescher PA, Müller-Eberhard HJ (eds):
on the antigenicity of macromolecules as determined by Textbook of Immunopathology, 2nd ed. Philadelphia: WB
radioimmunoassay (RIA), passive cutaneous anaphylaxis Saunders, 1976:421.
(PCA) and antibody induction. Int Archs Allergy Appl Immun 45. Miescher PA, Pola W: Haematological effects of non-narcotic
1977;55:23–28. analgesics. Drugs 1986;32(Suppl. 4):90.
20. Kitagawa M, Yagi Y, Pressman D: The heterogeneity of 46. Shulman NR: Immunoreactions involving platelets. I. A steric
combining sites of antibodies as determined by specific and kinetic model for formation of a complex from a human
immunoadsorbents. II. Comparison of elution patterns antibody, quinidine as a haptene, and platelets; and for fixation
obtained with anti-P-azobenzoate antibodies by different of complement by the complex. J Exp Med 1958;107:665.
kinds of immunoadsorbent and eluting hapten. J Immunol 47. Shulman NR, Rall JE: Mechanism of blood cell destruction in
1965;95:455–465. individuals sensitized to foreign antigens. Trans Assoc Amer
21. De Weck AL: Studies on penicillin hypersensitivity. I. The Physicians 1963;76:72.
specificity of rabbit “anti-penicillin” antibodies. Int Arch 48. Shulman N: Mechanism of blood cell damage by adsorption of
Allergy Appl Immunol 1962;21:20. antigen-antibody complexes. In Immunopathology, 3rd
22. De Weck AL: Newer developments in penicillin immuno- International Symposium, La Jolla, CA: Stuttgart: Schwabe,
chemistry. Int Arch Allergy Appl Immunol 1963;22:245. 1963:338.
23. Levine BB, Ovary Z: Studies on the mechanism of the forma- 49. Shulman NR: A mechanism of cell destruction in individuals
tion of the penicillin antigen. III. The N-(D-alpha-benzylpeni- sensitized to foreign antigens and its implications in autoim-
cilloyl) group as an antigenic determinant responsible for munity. Ann Int Med 1964;60:506.
hypersensitivity to penicillin. G J Exp Med 1961;114:875. 50. Neter E: Bacterial hemagglutination and hemolysis. Bacteriol
24. Levine BB, Price VH. Studies on the immunological mecha- Rev 1956;20:166–188.
nisms of penicillin allergy. II. Antigenic specificities of allergic 51. Boyden SV, Anderson ME: Agglutination of normal erythro-
wheal-and-flare skin responses in patients with histories of cytes in mixtures of antibody and antigen, and haemolysis in
penicillin allergy. Immunology 1964;7:542. the presence of complement. Br J Exp Pathol 1955;36:162–170.
25. Levine BB, Redmond AP: Minor haptenic determinant-specific 52. Miescher P, Cooper N: The fixation of soluble antigen-anti-
reagents of penicillin hypersensitivity in man. Int Arch Allergy body complexes upon thrombocytes. Vox Sang 1960;5:138–142.
Appl Immunol 1969;35:445–455. 53. Dameshek W: Autoimmunity: Theoretical aspects. Part 1. Ann
26. Parker CW, De Weck AL, Kern M, et al: The preparation and N Y Adac Sci 1965:124:6–28.
some properties of penicillenic acid derivatives relevant to 54. Cronin AE: The immunology of allergic drug reactions. PhD
penicillin hypersensitivity. J Exp Med 1962;15:803. Thesis, University of Cambridge, 1965.
27. Siegel BB, Levine BB: Antigenic specificities of skin-sensitizing 55. Salama A, Northoff H, Burkhardt H, et al: Carbimazole-
antibodies in sera from patients with immediate systemic induced immune haemolytic anaemia: Role of drug-red blood
allergic reactions to penicillin. J Allergy 1964;35:488. cell complexes for immunization. Brit J Haematol 1988;
28. Thiel JA, Mitchell S, Parker CW: Specificity of hemagglutina- 68:479–482.
tion reactions in human and experimental penicillin hypersen- 56. Christie DJ, Mullen PC, Aster RH: Fab-mediated binding of drug-
sitivity. J Allergy 1964;35:399. dependent antibodies to platelets in quinidine- and quinine-
29. Garratty G: Immune cytopenia associated with antibiotics. induced thrombocytopenia. J Clin Invest 1985;75:310–314.
Transfus Med Rev 1993;VII:255–267. 57. Smith ME, Reid DM, Jones CE, et al: Binding of quinine- and
30. Ackroyd JF: The pathogenesis of thrombocytopenic purpura quinidine-dependent drug antibodies to platelets is mediated
due to hypersensitivity to sedormid. Clin Sci 1949;7:249. by the Fab domain of the immunoglobulin G and is not Fc
31. Ackroyd JF: The immunological basis of purpura due to drug dependent. J Clin Invest 1987;79:912–917.
hypersensitivity. Proc Royal Soc Med 1962;55:30. 58. Jordan JV, Smith ME, Reid DM, et al: A tolmetin-dependent
32. Ackroyd JF, Rook AJ: Allergic drug reactions. In Gell PGH, antibody causing severe intravascular hemolysis binds to ery-
Coombs RRA (eds): Clinical Aspects of Immunology, 2nd ed. throcyte band 3 and requires only the F(ab′)2 domain to react
Oxford: Blackwell Scientific, 1968:693. [abstract]. Blood 1985;66(Suppl.):104a.
33. Ackroyd JF: Immunological mechanisms in drug hypersensi- 59. Shulman NR, Jones CE, Reid DM, et al: Evidence that drug,
tivity. In Gell PGH, Coombs RRA, Lachmann PJ (eds): Clinical not a neoantigen, is the primary determinant of drug-depend-
Aspects of Immunology, 3rd ed. Oxford: Blackwell Scientific, ent antibody reactions [abstract]. Blood 1993;82:268a.
1975:913. 60. Chong BH, Berndt MC, Koutts J, et al: Quinidine-induced
34. Ackroyd JF: Drug-induced thrombocytopenia. Vox Sang thrombocytopenia and leukopenia: Demonstration and char-
1983;45:257–259. acterization of distinct antiplatelet and anti-leukocyte antibo-
35. Ley AB, Harris JP, Brinkley M, et al: Circulating antibodies dies. Blood 1983;62:1218–1223.
directed against penicillin. Science 1958;127:1118. 61. Kunicki TJ, Johnson MM, Aster RH: Absence of the platelet-
36. Watson KC, Joubert SM, Bennett MAE: Occurrence of haemag- receptor for drug-dependent antibodies in the Bernard Soulier
glutinating antibody to penicillin. Immunology 1960;3:1. Syndrome. J Clin Invest 1978;62:716–719.
37. Levine BB, Redmond AP, Fellner MJ, et al: Penicillin allergy 62. van Leeuwen EF, Engelfriet CP, von dem Borne AEG Jr:
and the heterogeneous immune responses of man to ben- Studies on quinine- and quinidine-dependent antibodies
zylpenicillin. J Clin Invest 1966;45:1895–1906. against platelets and their reaction with platelets in the
38. Dewdney JM: Immunology of the antibiotics. In Sela M (ed): Bernard-Soulier syndrome. Brit J Haematol 1982;51:551–560.
The Antigens. New York: Academic Press, 1977:73–245. 63. Kunicki TJ, Russell N, Nurden AT, et al: Further studies of the
39. Shaltiel S, Mizrahi R, Sela M: On the immunological properties human platelet receptor for quinine- and quinidine-dependent
of penicillins. Proc R Soc Lond B 1971;179:411–432. antibodies. J Immunol 1981;126:398–402.
40. Neftel KA: Effect of storage of penicillin-G solutions on sensi- 64. Berndt MC, Chong BH, Bull HA, et al: Molecular characterization
tisation to penicillin-G after intravenous administration. of quinine/quinidine drug-dependent antibody platelet inter-
Lancet 1982;1:986–988. action using monoclonal antibodies. Blood 1985;66:1292–1301.
41. Neftel KA, Walti M, Schulthess HK, et al: Adverse reactions 65. Devine DV, Rosse WF: Identification of platelet proteins that bind
following intravenous penicillin-G relate to degradation of the alloantibodies and autoantibodies. Blood 1984;64:1240–1245.
drug in vitro. Klin Wochenschr 1984;62:25–29. 66. Christie DJ, Mullen PC, Aster RH: Quinine- and quinidine
42. Miescher PA, Miescher A: Die sedormid-anaphylaxie. Schweiz platelet antibodies can react with GPIIb/IIIa. Brit J Haematol
Med Wochenschr 1952;82:1279. 1987;67:213–219.
43. Miescher PA, Gorstein F: Mechanisms of immunogenic 67. Visentin GP, Newman PJ, Aster RH: Characteristics of quinine-
platelet damage. In Johnson SA, Monto RW, Rebuck JW, Horn and quinidine-induced antibodies specific for platelet glyco-
RC (eds): Blood Platelets. London: J & A Churchill, 1961:671. proteins IIb and IIIa. Blood 1991;77:2668–2676.
312 Immune Hemolytic Anemias

68. Christie DJ, Aster RH: Drug-antibody-platelet interaction in of auto- and/or drug-dependent antibodies against these cells.
quinine- and quinidine-induced thrombocytopenia. J Clin Br J Haematol 1987;66:263–266.
Invest 1982;70:989–998. 94. Salama A, Göttsche B, Mueller-Eckhardt C: Autoantibodies
69. Claas FHJ, Langerak J, van Rood JJ: Drug-induced antibodies and drug- or metabolite-dependent antibodies in patients with
with restricted specificity. Immunol Letters 1981;2:323. diclofenac-induced immune haemolysis. Br J Haematol
70. Letona J, Barbolla L, Frieyro E, et al: Immune haemolytic 1991;77:546–549.
anaemia and renal failure induced by streptomycin. Br J 95. Squires JE, Mintz PD, Clark S: Tolmetin-induced hemolysis.
Haematol 1977;35:561–571. Transfusion 1985;25:410–413.
71. Duran-Suarez JR, Martin-Vega C, Argelagues E, et al: Red cell 96. van Dijk BA, Rico PB, Hoitsma A, et al: Immune hemolytic
I antigen as immune complex receptor in drug-induced anemia associated with tolmetin and suprofen. Transfusion
hemolytic anemias. Vox Sang 1981;41:313–315. 1989;29:638–641.
72. Habibi B, Basty R, Chodez S, et al: Thiopental-related immune 97. Schulenburg BJ, Beck ML, Pierce SR, et al: Immune hemolysis
hemolytic anemia and renal failure. N Engl J Med associated with ZomaxT [abstract]. Transfusion 1983;23:409.
1985;312:353–355. 98. Toy E, Nesbitt R, Savastano G, et al: Warm autoantibody fol-
73. Salama A, Mueller-Eckhardt C: On the mechanisms of sensiti- lowing plasma apheresis, complicated by acute intravascular
zation and attachment of antibodies to RBC in drug-induced hemolysis associated with cefoxitin-dependent antibody
immune hemolytic anemia. Blood 1987;69:1006–1010. resulting in fatality [abstract]. Transfusion 1989;29:51S.
74. Sandvei P, Nordhagen R, Michaelsen TE, et al: Fluorouracil 99. Weitekamp LA, Johnson ST, Fueger JT, et al: Cefotetan-
(5-FU) induced acute immune haemolytic anaemia. Br J dependent immune hemolytic anemia due to a single antibody
Haematol 1987;65:357–359. reacting with both drug coated and untreated red cells in the
75. Pereira A, Sanz C, Cervantes F, et al: Immune hemolytic anemia presence of drug. In Book of Book of Abstracts. ISBT/AABB
and renal failure associated with rifampicin-dependent anti- Joint Congress. Arlington, VA: American Association of Blood
bodies with anti-I specificity. Ann Hematol 1991;63:56–58. Banks, 1990:33.
76. Habibi B, Bretagne Y: Blood group antigens may be the recep- 100. Wojcicki RE, Larson CJ, Pope ME, et al: (1990). Acute
tors for specific drug-antibody complexes reacting with red hemolytic anemia during cefotetan therapy. Book of Abstracts.
blood cells. CR Acad Sc (D) (Paris) 1983;296:693. ISBT/AABB Joint Congress. Arlington, VA: American
77. Sosler SD, Behzad O, Garratty G, et al: Acute hemolytic anemia Association of Blood Banks, 1990:33.
associated with a chlorpropamide-induced apparent auto anti- 101. Chambers LA, Donovan LM, Kruskall MS: Ceftazidime-
Jka. Transfusion 1984;24:206–209. induced hemolysis in a patient with drug-dependent antibo-
78. Beck ML: A fatty-acid dependent antibody with Rh specificity. dies reactive by immune complex and drug adsorption
In Book of Abstracts, ISBT/AABB Joint Congress. Arlington, mechanisms. Am J Clin Pathol 1991;95:393–396.
VA: American Association of Blood Banks, 1972:6. 102. Gallagher MT, Schergen AK, Sokol-Anderson ML, et al: Severe
79. Dube VE, Zoes C, Adesman P: Caprylate-dependent autoanti- immune mediated hemolytic anemia secondary to treatment
e.Vox Sang 1977;33:359–363. with cefotetan. Transfusion 1992;32:266–268.
80. Reviron M, Janvier D, Reviron J, et al: An anti-I cold autoag- 103. Garrratty G: Drug-induced immune hemolytic anemia. In
glutinin enhanced in the presence of sodium azide. Vox Sang Garratty G (ed): Immunobiology of transfusion medicine.
1984;46:211–216. New York: Dekker, 1994:523–551.
81. Halima D, Garratty G, Bueno R: An apparent anti-Jka reacting 104. Arndt PA, Leger RM, Garratty G: Serology of antibodies to
only in the presence of methyl esters of hydroxybenzoic acid. second- and third-generation cephalosporins associated with
Transfusion 1982;22:521–524. immune hemolytic anemia and/or positive direct antiglobulin
82. Judd WJ, Steiner EA, Cochran RK: Paraben-associated tests. Transfusion 1999;39:11239–11246.
autoanti-Jka antibodies. Transfusion 1982;22:31–35. 105. Worlledge SM, Carstairs KC, Dacie JV: Autoimmune
83. Muirhead EE, Groves M, Guy R, et al: Acquired hemolytic haemolytic anaemia associated with alpha-methyldopa
anemia, exposure to insecticides and positive Coombs test therapy. Lancet 1966;2:135–139.
dependent on insecticide preparations. Vox Sang 1959;4:277. 106. Weigle WO: The induction of autoimmunity in rabbits follow-
84. Hart MN, Mesara BW: Phenacetin antibody cross-reactive ing injection of heterologous or altered homologous thy-
with autoimmune erythrocyte antibody. Am J Clin Pathol roglobulin. J Exp Med 1965;121:289–308.
1969;52:695–701. 107. Weigle WO: The antibody response in rabbits to previously
85. Garratty G, Houston M, Petz LD, et al: Acute immune tolerated antigens. Ann N Y Acad Sci 1965;124:133–142.
intravascular hemolysis due to hydrochlorothiazide. Am J Clin 108. Worlledge SM: Autoantibody formation associated with
Pathol 1981;76:73–78. methyldopa (Aldomet) therapy. Br J Haematol 1979;15:5–8.
86. Shulman IA, Arndt PA, McGehee W: Cefotaxime-induced 109. Green FA, Jung CY, Rampal A, et al: Alpha-methyldopa and the
immune hemolytic anemia due to antibodies reacting in vitro erythrocyte membrane. Clin Exp Immunol 1980;40:554–560.
by more than one mechanism. Transfusion 1990;30:263–266. 110. Green FA, Jung CY, Hui H: Modulation of alphamethyldopa
87. Garratty G, Nance S, Lloyd M, et al: Fatal immune hemolytic binding to the erythrocyte membrane by superoxide dismu-
anemia due to cefotetan. Transfusion 1992;32:269–271. tase. Biochem Biophys Res Comm 1980;95:1037–1042.
88. Shirey RS, Morton SJ, Lawton KB, et al: Fenoprofen-induced 111. LoBuglio AF, Jandl JH: The nature of the alphamethyldopa
immune hemolysis: Difficulties in diagnosis and complica- red-cell antibody. N Engl J Med 1967;276:658–665.
tions in compatibility testing. Am J Clin Pathol 1998;89: 112. Sherman JD, Love DE, Harrington JF: Anemia, positive lupus
410–414. and rheumatoid factors with methyldopa. Arch Intern Med
89. Florendo NT, MacFarland D, Painter M, et al: Streptomycin- 1967;120:321–326.
specific antibody coincident with a developing warm autoan- 113. Harm M: LE cells and positive direct Coombs’ test induced by
tibody. Transfusion 1980;20:662–668. methyldopa. Can Med Assoc J 68;99:277–280.
90. Habibi B, Lopez M, Serdaru M, et al: Immune hemolytic 114. Mackay IR, Cowling DC, Hurley TH: Drug-induced autoim-
anemia and renal failure due to teniposide. N Eng J Med mune disease: Haemolytic anaemia and lupus cells after treat-
1982;306:1091–1093. ment with methyldopa. Med J Aust 1968;2:1047–1050.
91. Bird GWG, Wingham J, Babb RG, et al: (1984). Azapropazone- 115. Devereux S, Fisher DM, Foter BLT, et al: Factor VIII inhibitor
associated antibodies. Vox Sang 1984;46:336–337. and raised platelet IgG levels associatred with methyldopa
92. Salama A, Mueller-Eckhardt C: Two types of nomifensine- therapy. Br J Haematol 1983;54:485–488.
induced immune haemolytic anaemias: Drug-dependent 116. Feltkamp TEW, Engelfriet CP, Van Loghem JJ: Autoantibodies
sensitization and/or autoimmunization. Br J Haematol 1986; and methyldopa. Lancet 1968;1:644.
64:613–620. 117. Perry HM, Chaplin H, Carmody S, et al: Immunologic findings
93. Salama A, Mueller-Eckhardt C: Cianidanol and its metabolites in patients receiving methyldopa: A prospective study. J Lab
bind tightly to red cells and are responsible for the production Clin Med 1971;78:905–917.
Drug-Induced Immune Hemolytic Anemia 313

118. Habibi B: Drug induced red blood cell autoantibodies co- 144. Levine BB, Redmond A: Immunochemical mechanisms of
developed with drug specific antibodies causing haemolytic penicillin induced Coombs positivity and hemolytic anemia in
anaemias. Br J Haematol 1985;61:139–143. man. Int Arch Allergy Appl Immunol 1967;31:594–606.
119. Mueller-Eckhardt C, Salama A: Drug-induced immune 145. White JM, Brown DL, Hepner GW, et al: Penicillin-induced
cytopenias: A unifying pathogenetic concept with special haemolytic anaemia. Br Med J 1968;3:26–29.
emphasis on the role of drug metabolites. Transfus Med Rev 146. Kerr RO, Cardamone J, Dalmasso AP, et al: Two mechanisms
1990;4:69–77. of erythrocyte destruction in penicillin-induced hemolytic
120. Garratty G: Target antigens for red-cell-bound autoantibodies. anemia. N Eng J Med 1972;287:1322–1325.
In Nance SJ (ed). Clinical and basic science aspects of immuno- 147. Reis CA, Rosenbaum TJ, Garratty G, et al: Penicillin-induced
hematology. Arlington, VA: American Association of Blood immune hemolytic anemia. JAMA 1975;233:432–435.
Banks, 1991:33–72. 148. Funicella T, Weinger RS, Moake JL, et al: Penicillin-induced
121. Dameshek W: Alpha-methyldopa red-cell antibody: Cross- immunohemolytic anemia associated with circulating immune
reaction or forbidden clones. N Engl J Med 1967;276:1382. complexes. Am J Hematol 1977;3:219–223.
122. Petz LD, Fudenberg HH: Immunologic mechanisms in drug- 149. Molthan L, Reidenberg MM, Eichman MF: Positive direct
induced cytopenias. Prog Hematol 1975;9:185–206. Coombs tests due to cephalothin. N Engl J Med 1967;
123. Kirtland HH, Mohler DN, Horwitz DA: Methyldopa 277:123–125.
inhibition of suppressor-lymphocyte function. N Engl J Med 150. Gralnick HR, Wright LD Jr, McGinniss MH: Coombs’ positive
1980;302:825–832. reactions associated with sodium cephalothin therapy. JAMA
124. Lipsky PE, Ginsburg WW, Finkelman FD, et al: Control of human 1967;199:135–136.
B lymphocyte responsiveness: Enhanced suppressor T cell 151. Perkins RL, Mengel CE, et al: Direct Coombs’ test reactivity
activity after in vitro incubation. J Immunol 1978;120:902–910. after cephalothin or cephaloridine in man and monkey. Proc
125. Garratty G, Arndt P, Prince HP, et al: The effect of methyldopa Soc Exptl Biol Med 1968;129:397–401.
and procainamide on suppressor cell activity. Br J Haematol 152. Spath P, Garratty G, Petz LD: Studies on the immune response
1992;84:310–315. to penicillin and cephalothin in humans. II. Immunohema-
126. Kleinman S, Nelson R, Smith L, et al: Positive direct antiglo- tologic reactions to cephalothin administration. J Immunol
bulin tests and immune hemolytic anemia in patients receiv- 1971;107:860–869.
ing procainamide. N Engl J Med 1984;311:809–812. 153. Spath P, Garratty G, Petz L: Studies on the immune response to
127. Rubin RL: Autoimmune reactions induced by procainamide and penicillin and cephalothin in humans. I. Optimal conditions
hydralazine. In Kammüller ME, Bloksma N, Seinen W (eds): for titration of hemagglutinating penicillin and cephalothin
Autoimmunity and Toxicology. New York: Elsevier, 1989:119. antibodies. J Immunol 1971;107:854–859.
128. Blomgren SE, Condemi JJ, Vaughan JH: Procainamide-induced 154. Branch DR, Sy Siok Hian AL, Petz LD: Mechanism of nonim-
lupus erythematosus: Clinical and laboratory observations, munologic adsorption of proteins using cephalothin-coated
Am J Med 1972;52:338–348. red cells [abstract]. Transfusion 1985;24:415.
129. Gold EF, Ben-Efraim S, Faivisewitz A, et al: Experimental 155. Garratty G, Leger R: Red cell membrane proteins (CD55 and
studies on the mechanism of induction of anti-nuclear anti- CD58) are modified following treatment of RBCs with
bodies by procainamide. Clin Immunol Immunopathol cephalosporins [abstract]. Blood 1995;86:68a.
1977;7:176–186. 156. Sirchia G, Murcuriali F, Ferrone S: Cephalothin-treated normal
130. Schoen RT, Trentham DE: Drug-induced lupus: An adjuvant red cells: A new type of PNH-like cells. Experientia
disease? Am J Med 1981;71:5–8. 1968;24:495–496.
131. Cornacchia E, Golbus J, Maybaum J, et al: Hydralazine and 157. Ferrone S, Zanella A, Mercuriali F, et al: Some enzymatic and
procainamide inhibit T cell DNA methylation and induce metabolic activities of normal human erythrocytes treated in
autoreactivity. J Immunol 1988;140:2197–2200. vitro with cephalothin. Eur J Pharmacol 1968;4:211–214.
132. Bluestein HG, Weisman MH, Zvaifler N, et al: Lymphocyte 158. Rosse WF: Phosphatidylinositol-linked proteins and paroxys-
alteration by procainamide: Relation to drug-induced lupus mal nocturnal hemoglobinuria. Blood 1990;75:1595–1601.
erythematosus syndrome. Lancet 1979;2:816–819. 159. Jamin D, Demers J, Shulman I, et al: An explanation for non-
133. Bluestein HG, Redelman D, Zvaifler NJ: Procainamide- immunologic adsorption of proteins onto red blood cells.
lymphocyte reactions. Arthr Rheum 1981;24:1019–1023. Blood 1986;67:993–996.
134. Miller KB, Salem D: Immune regulatory abnormalties pro- 160. Jamin D, Shulman I, Lam HT, et al: Production of a positive
duced by procainamide. Am J Med 1982;73:487–492. direct antiglobulin test due to Suramin. Arch Pathol Lab Med
135. DeBoccardo G, Drayer D, Rubin AL, et al: Inhibition of poke- 1986;112:898–900.
weed mitogen-induced B cell differentiation by compounds 161. Zeger G, Smith L, McQuiston D, et al: Cisplatin-induced non-
containing primary amine or hydrazine groups. Clin Exp immunologic adsorption of immunoglobulin by red cells.
Immunol 1985;59:69–76. Transfusion 1988;28:493–495.
136. Yu C-L, Ziff M: Effects of long-term procainamide therapy on 162. Getaz EP, Beckley S, Fitzpatrick, J, et al: Cisplatin-induced
immunoglobulin synthesis. Arthr Rheum 1985;28:276–284. hemolysis. N Engl J Med 1980;302:334–335.
137. Ochi T, Goldings EA, Lipsky PE, et al: Immunomodulatory 163. Levi JA, Aroney RS, Dalley DN: Haemolytic anaemia after cis-
effect of procainamide in man. J Clin Invest 1983;71:36–45. platin treatment. Br Med J 1981;282:2003–2004.
138. Seeman P: The membrane actions of anesthetics and tranquil- 164. Nguyen BV, Lichtiger B: Cisplatin-induced anemia. Cancer
izers. Pharmacol Rev 1972;24:583–655. Treat Rep 1981;65:1121.
139. Shulman NR, Reid DM: Mechanisms of drug-induced 165. Cinollo G, Dini G, Franchini E, et al: Positive direct antiglobu-
immunologically mediated cytopenias. Transfus Med Rev lin tests in a pediatric patient following high-dose cisplatin.
1993;VII:215–229. Cancer Chemother Pharmacol 1988;21:85–86.
140. Garratty G, Petz LD: Drug-induced immune hemolytic 166. Weber JC, Couppie P, Maloisel F, et al: Anémie hémolytique au
anemia. Am J Med 1975;58:398–407. cisdiamino-dichloroplatinum. La Presse Médicale 1990;19:
141. Petz LD, Fudenberg HH: Coombs-positive hemolytic anemia 526–527.
caused by penicillin administration. N Engl J Med 1966; 167. Marani TM, Trich MB, Armstrong KS, et al: Carboplatin-
274:171–178. induced immune hemolytic anemia. Transfusion 1996;36:
142. Abraham GN, Petz LD, Fudenberg HH: Immunohaematolo- 1016–1018.
gical cross-allergenicity between penicillin and cephalothin in 168. Desrame J, Broustet H, de Talilly PD, et al: Oxaliplatin-induced
humans. Clin Exptl Immunol 1968;3:343–357. haemolytic anaemia. Lancet 1999;354:1179–1180.
143. Croft JD, Swisher SN, Gilliland BC, et al: Coombs-test 169. Garufi C, Vaglio S, Brienza S, et al: Immunohemolytic anemia
positivity induced by drugs: Mechanisms of immuno- following oxaliplatin administration. Ann Oncol 2000;11:497.
logic reactions and red cell destruction. Ann Intern Med 170. Earle CC, Chen WY, Ryan DP, et al: Oxaliplatin-induced
1968;68:176–187. Evan’s syndrome. Br J Cancer 2001;84:441.
314 Immune Hemolytic Anemias

171. Sørbye H, Bruserud Ø, Dahl O: Oxaliplatin-induced haemato- 197. Orenstein AA, Yakulis V, Eipe J, et al: Immune hemolysis due
logical emergency with an immediate severe thrombocytope- to hydralazine. Ann Int Med 1977;86:450–451.
nia and haemolysis. Acta Oncol 2001;40:882–883. 198. Petersen BH, Graham J: Immunologic cross-reactivity of
172. Arndt P, Garratty G: Positive direct antiglobulin tests associ- cephalexin and penicillin. J Lab Clin Med 1974;83:860–870.
ated with oxaliplatin can be caused by antibody and/or 199. van der Meulen FW, de Bruin HG, Goosen PCM, et al:
nonimmunological protein adsorption. Transfusion 2003 (in Quantitative aspects of the destruction of red cells sensitized
press). with IgG1 autoantibodies: An application of flow cytofluorom-
173. Lutz P, Dzik W: Very high incidence of a positive direct etry. Br J Haematol 1980;46:47–56.
antiglobulin test (+DAT) in patients receiving Unasyn® 200. Garratty G, Nance S: Correlation between in vivo hemolysis
[abstract]. Transfusion 1992;32:23S. and the amount of red cell-bound IgG measured by flow
174. Garratty G, Arndt PA: Positive direct antiglobulin tests and cytometry. Transfusion 1990;30:617–621.
haemolytic anaemia following therapy with beta-lactamase 201. Carstairs KC, Breckenridge A, Dollery CT, et al: Incidence of a
inhibitor containing drugs may be associated with nonim- positive direct Coombs test in patients on α-methyldopa.
munologic adsorption of protein onto red blood cells. Br J Lancet 1966;ii:133–135.
Haematol 1998;100:777–783. 202. Hunter E, Raik E, Gordon S, et al: Incidence of positive Coombs’
175. Williams ME, Thomas D, Harman CP, et al: Positive direct test, LE cells and antinuclear factor in patients on alpha-methyl-
antiglobulin tests due to clavulanic acid. Antimicrob Agents dopa (Aldomet) therapy. Med J Aust 1971;2:810–812.
Chemother 1985;27:125–127. 203. Dollery CT, Worlledge SM, Holbrow EJ, et al: Positive direct
176. Broadberry RE, Farren TW, Kohler JA, et al: Haemolytic Coombs tests and antinuclear factor in patients treated with
anaemia associated with tazobactam [abstract]. Vox Sang methyldopa. Lancet 1967;2:1265–1268.
2002;83(S2):227. 204. Woodgate D, Feizi T: The direct antiglobulin (Coombs’) test in
177. Arndt P, Garratty G: Can nonspecific protein coating of RBCs hypertensive patients. Vox Sang 1967;12:273–278.
lead to increased RBC destruction? Drug-treated RBCs with 205. Worlledge SM: Immune drug-induced hemolytic anemias.
positive antiglobulin tests due to nonspecific protein uptake Semin Hematol 1973;10:327–344.
give positive monocyte monolayer assays [abstract]. 206. Worlledge SM: Autoantibody formation associated with
Transfusion 2000;40:29S. methyldopa (Aldomet) therapy. Br J Haematol 1979;16:5–8.
178. Nance SJ, Arndt P, Garratty G: Predicting the clinical 207. Bakemeier RF, Leddy JP: Erythrocyte autoantibody associated
significance of red cell alloantibodies using a monocyte mono- with alpha methyldopa: Heterogeneity of structure and
layer assay. Transfusion 1987;27:449–452. specificity. Blood 1968;32:1–14.
179. Garratty G: Predicting the clinical significance of red cell 208. Lalezari P, Louie JE, Fadlallah N: Serologic profile of
antibodies with in vitro cellular assays. Transfus Med Rev alphamethyldopa-induced hemolytic anemia: Correlation
1990;IV:297–312. between cell-bound IgM and hemolysis. Blood 1982;59:61–68.
180. Ley AB, Cahan A, Mayer K: Circulating antibody directed 209. LoBuglio AF, Jandl JH: The nature of the alpha methyldopa
against penicillin. Bibl Haemat 1959;10:539. red-cell antibody. N Engl J Med1967;276:658–665.
181. Petz LD: Immunologic reaction of humans to cephalosporins. 210. Issitt PD, Pavone BG, Goldfinger D, et al: Anti-Wrb and other
Postgraduate Med J 1971;47(Suppl.):64–69. autoantibodies responsible for positive direct antiglobulin test
182. Levine BB: Immunologic mechanisms of penicillin allergy: A in 150 individuals. Br J Haematol 1976;34:5–18.
haptenic model system for the study of allergic diseases of 211. Patten E, Beck CE, Scholl C, et al: Autoimmune hemolytic
man. N Engl J Med 1966;275:1115–1125. anemia with anti-Jka specificity in a patient taking Aldomet.
183. Gralnick HR, McGinnis M, Elton W, et al: Hemolytic anemia Transfusion 1977;17:517–520.
associated with cephalothin. JAMA 1971;217:1193–1197. 212. Kessey EC, Pierce S, Beck ML, et al: Alpha-methyldopa-
184. Jeannet M, Bloch A, Dayer JM, et al: Cephalothin-induced induced hemolytic anemia involving autoantibody with U
immune hemolytic anemia. Acta Haemat 1976;55:109–117. specificity [abstract]. Transfusion 1973;13:360.
185. Rubin RN, Burka ER: Anti-cephalothin antibody and 213. Branch DR: Gallagher MT, Shulman IA, et al: Reticulo-
Coombs’-positive hemolytic anemia. Ann Int Med 1977;86: endothelial cell function in α-methyldopa-induced hemolytic
64–65. anemia. Vox Sang 1983;45:278–287.
186. Moake JL, Butler CF, Hewell GM, et al: Hemolysis induced by 214. Gallagher MT, Branch DR, Mison A, et al: Evaluation of reticu-
cefazolin and cephalothin in a patient with penicillin sensitiv- loendothelial function in autoimmune hemolytic anemia using
ity. Transfusion 1978;18:369–373. an in vitro assay of monocyte-macrophage interaction with
187. Branch DR, Berkowitz LR, Becker RL, et al: Extravascular erythrocytes. Exp Hematol 1983;11:82–89.
hemolysis following the administration of cefamandole. Am J 215. Yust I, Frisch B, Goldsher N: Antibody-dependent cell-
Hematol 1985;18:213–219. mediated cytotoxicity and phagocytosis of autologous red
188. Thomson S, Williamson D: A case of ampicillin-induced blood cells in alphamethyldopa-induced haemolysis. Scand J
haemolytic anemia. Canad J Med Tech 1974;36:228–229. Haematol 1986;36:211–216.
189. Bell CA, Zwicker H, Whitcomb M: Matuhasi-Ogata phenome- 216. Kelton JG: Impaired reticuloendothelial function in patients
non involving anti-ampicillin. Transfusion 1978;18:244–249. treated with methyldopa. N Engl J Med 1985;313:596–600.
190. Gmür J, Wälti M, Neftel KA: Amoxicillin-induced immune 217. Worlledge S: Immune drug induced haemolytic anaemias. In
hemolysis. Acta Haemat 1985;74:230–233. Girdwood RH (ed): Blood disorders due to drugs and other
191. Johnson ST, Weitekamp LA, Sauer DE, et al: Piperacillin- agents. Amsterdam: Excerpta Medica, 1973:11–26.
dependent antibody with relative e specificity reacting with 218. Petz LD: Drug-induced immune haemolytic anaemia. Clin
drug treated red cells and untreated red cells in the presence of Haematol 1980;9:455–482.
drug [abstract]. Transfusion 1994;34:20S. 219. Silvergleid AJ, Wells RF, Hafleigh EB, et al: Compatibility test
192. Arndt PA, Garratty G, Hill J, et al: Two cases of immune using 51chromium-labelled red blood cells in crossmatch
haemolytic anaemia, associated with anti-piperacillin, positive patients. Transfusion 1978;18:8–14.
detected by the “immune complex” method. Vox Sang 2002; 220. Snyder EL, Spivak M: Clinical and serological management of
83:273–278. patients with methyldopa-induced positive antiglobulin tests.
193. Seldon MR, Bain B, Johnson CA, et al: Ticarcillin-induced im- Transfusion 1979;19:313–316.
mune haemolytic anaemia. Scand J Haematol 1982;28:459–460. 221. Parr GVS, Ballard JO: Autologous blood transfusion with
194. Arndt PA, Wolf CF, Kripas CT, et al: First example of an anti- methyldopa induced positive direct antiglobulin test.
body to ticarcillin: A possible cause of hemolytic anemia Transfusion 1980;20:119.
[abstract]. Transfusion 1999;39:47S. 222. Bastion Y, Coiffier B, Dumontet C, et al: Severe autoimmune
195. Kroovand S, Kirtland H, Issitt C: A positive direct antiglobulin hemolytic anemia in two patients treated with fludarabine for
test due to sodium nafcillin [abstract]. Transfusion 1977;17:682. chronic lymphocytic leukemia. Ann Oncol 1992;3:171–172.
196. Garratty G, Brunt D: Difficulties in detecting nafcillin antibo- 223. Tosti S, Caruso R, D’Adamo F, et al: Severe autoimmune
dies [abstract]. Transfusion 1983;23:409. hemolytic anemia in a patient with chronic lymphocytic
Drug-Induced Immune Hemolytic Anemia 315

leukemia responsive to fludarabine-based treatment. Ann 247. Henry RE, Goldberg LS, Sturgeon P, et al: Serologic abnormal-
Hematol 1992;65:238–239. ities associated with L-dopa therapy. Vox Sang 1971;20:
224. Di Raimondo F, Giustolisi R, Cacciola E, et al: Autoimmune 306–316.
hemolytic anemia in chronic lymphocytic leukemia patients 248. Joseph C: Occurrence of positive Coombs test in patients
treated with fludarabine. Leuk Lymph 1993;11:63–68. treated with levodopa. N Engl J Med 1972;286:1401–1402.
225. Byrd JC, Hertler AA, Weiss RB, et al: Fatal recurrence of 249. Gabor EP, Goldberg LS: Levodopa induced Coombs positive
autoimmune anemia following pentostatin therapy in a haemolytic anaemia. Scand J Haematol 1973;11:201–203.
patient with a history of fludarabine-associated hemolytic 250. Lindstrom FD, Lieden G, Engstrom MS: Dose-related
anemia. Ann Oncol 1995;6:730–731. levodopa-induced haemolytic anaemia. Ann Intern Med
226. Myint H, Cooplestone JA, Orchard J, et al: Fludarabine-related 1977;86:298–300.
autoimmune haemolytic anaemia in patients with chronic 251. Bernstein RM: Reversible haemolytic anaemia after levodopa-
lymphocyte leukaemia. Br J Haematol 1995;91:341–344. carbidopa. Br Med J 1979;1:1461–1462.
227. Tertian G, Cartron J, Bayle C, et al: Fatal intravascular autoim- 252. Territo MC, Peters RW, Tanaka KR: Autoimmune hemolytic
mune hemolytic anemia after fludarabine treatment for chronic anemia due to levodopa therapy. JAMA 1973;226:1347–1348.
lymphocyte leukemia. Hematol Clin Ther 1996;38:359–360. 253. Jones GW, George TL, Bradley RD: Procainamide-induced
228. Maclean R, Meiklejohn D, Soutar R: Fludarabine-related hemolytic anemia. Transfusion 1978;18:224–227.
autoimmune haemolytic anaemia in patients with chronic 254. Kornberg A, Rachmilewitz E: Procainamide-induced hemoly-
lymphocytic leukaemia. Br J Haematol 1996;92:766–773. tic anemia in a patient with traumatic cardiac hemolytic
229. Robak T, Blasiska-Morawiec M, Krykowski E, et al: anemia. Arch Intern Med 1981;141:1388.
Autoimmune haemolytic anaemia in patients with chronic 255. Schifman RB, Garewal H, Shillington D: Reticulocytopenic.
lymphocytic leukaemia treated with 2-chlorodeoxyadenosine Coombs’ positive anemia induced by procainamide. Am J Clin
(cladribine). Eur J Haematol 1997;58:109–113. Pathol 1983;80:66–68.
230. Longo G: Fludarabine and autoimmune hemolytic anemia in 256. Kleinman S, Nelson R, Smith L, et al: Positive direct antiglo-
chronic lymphocytic leukemia. Eur J Haematol 1997;59: bulin tests and immune hemolytic anemia in patients receiv-
124–125. ing procainamide. N Engl J Med 1984;311:809–812.
231. Chasty RC, Myint H, Oscier DG, et al: Autoimmune haemoly- 257. Sanford-Driscoll M, Knodel LC: Induction of hemolytic
sis in patients with B-CLL treated with chlorodeoxtyadenosine anemia by nonsteroidal antiinflammatory drugs. Drug Intel
(CDA). Leuk Lymph 1998;29:391–398. Clin Pharm 1986;20:925–934.
232. Vick DJ, Byrd JC, Beal CL, et al: Mixed-typed autoimmune 258. Kramer MR, Levene C, Hershko C: Severe reversible autoim-
hemolytic anemia following fludarabine treatment in a patient mune haemolytic anaemia and thrombocytopenia associated
with chronic lymphocytic leukemia/small cell lymphoma. Vox with diclofenac therapy. Scand J Haematol 1986;36:118–120.
Sang 1998;74:122–126. 259. Chan-Lam D, Thorburn AW, Chalmers EA, et al: Red cell anti-
233. Gonzalez H, Leblond V, Azar N, et al: Severe autoimmune bodies and autoimmune haemolysis after treatment with
hemolytic anemia in eight patients treated with fludarabine. azapropazone. Br Med J 1986;293:1474.
Hematol Cell Ther 1998;40:113–118. 260. Salama A, Göttsche B, Mueller-Eckhardt C: Autoantibodies
234. Weiss RB, Freiman J, Kweder SL, et al: Hemolytic anemia after and drug- or metabolite-dependent antibodies in patients with
fludarabine therapy for chronic lymphocytic leukemia. J Clin diclofenac-induced immune haemolysis. Br J Haematol
Oncol 1998;16:1885–1889. 1991;77:546–549.
235. Hamblin TJ, Orchard JA, Myint H, et al: Fludarabine and 261. Johnson ST, Fueger JT, Gottschall JL, et al: Ibuprofen-dependent
hemolytic anemia in chronic lymphocytic leukemia. J Clin antibody with Rh specificity reacting with untreated red cells in
Oncol 1998;16:3209–3210. the presence of drug. Transfusion 1996;36:54S.
236. Tetreault SA, Saven A: Delayed onset of autoimmune hemolytic 262. Laidlaw ST, Stamps R, Booker DJ, et al: Immune hemolytic
anemia complicating cladribine therapy for Waldenstrom anemia due to dicofenac. Immunohematology 1997;13:9–11.
macroglobulinemia. Leuk Lymph 2000;37:125–130. 263. Bougie D, Johnson ST, Weitekamp LA, et al: Sensitivity to a
237. Mauro FR, Foa R, Cerretti R, et al: Autoimmune hemolytic metabolite of diclofenac as a cause of acute immune hemolytic
anemia in chronic lymphocytic leukemia: Clinical, therapeutic, anemia. Blood 1997;90:407–413.
and prognostic features. Blood 2000;95:2786–2792. 264. Madoz P, Muñiz-Diaz E, Martinez C, et al: Fatal immune
238. Dighiero G, Travade P, Chevret S, et al: B-cell chronic lympho- hemolytic anemia induced by aceclofenac [abstract]. Trans-
cytic leukemia: Present status and future directions. Blood fusion 1997;37:36S.
1991;78:1901–1914. 265. van Dijk BA, Rico PB, Hoitsma A, et al: Immune hemolytic
239. Bersagel DE: The chronic leukemias: A review of disease anemia associated with tolmetin and suprofen. Transfusion
manifestations and the aim of therapy. Canadian Med Assoc J 1989;29:638–641.
1967;96:1615–1620. 266. Weitekamp LA, Johnson ST, Fueger JT, et al: A fatal case of tol-
240. Hamblin TJ, Oscier DG, Young BJ: Autoimmunity in chronic metin-induced hemolytic anemia with relative e specificity
lymphocytic leukemia. J Clin Pathol 1986;39:713–716. [abstract]. Transfusion 1991;31:52S.
241. Astrow AB: Fludarabine in chronic leukemia. Lancet 1996; 267. McCall L, Owens M: Case report: Hemolytic anemia produced
347:1420–1421. by tolmetin. Immunohematology 1992;8:17–18.
242. Diehl LF, Ketchum LH: Autoimmune disease and chronic lym- 268. Weitekamp LA, Johnson ST, Fueger JT, et al: Sulindac-induced
phocytic leukemia: Autoimmune hemolytic anemia, pure red hemolytic anemia caused by antibody reacting with untreated
cell aplasia, and autoimmune thrombocytopenia. Semin Oncol red cells in the presence of drug [abstract]. Transfusion
1998;25:80–97. 1992;32:54S.
243. Johnson S, Smith AG, Loffler H, et al: Multicentre prospective 269. DeCoteau J, Reis MD, Pinkerton PH, et al: Positive antiglobu-
randomized trial of fludarabine versus cyclophosphamide, lin test in association with sulindac: Involvement of the Rh
doxorubicin, and prednisone (CAP) for treatment of factor. Vox Sang 1993;64:179–183.
advanced-stage chronic lymphocytic leukaemia. The French 270. Angeles ML, Reid ME, Yacob UA, et al: Sulindac-induced
Cooperative Group on CLL: Lancet 1996;347:1432–1438. immune hemolytic anemia. Transfusion 1994;34:255–258.
244. Orchard J, Bolam S, Myint H, et al: In patients with lymphoic 271. Cunha PD, Lord RS, Johnson ST, et al: Immune hemolytic
tumours recovering from the autoimmune complications of anemia caused by sensitivity to a metabolite of etodolac, a non-
fludarabine, relapse may be triggered by conventional steroidal anti-inflammatory drug. Transfusion 2000;40:663–668.
chemotherapy. Br J Haematol 1998;102:1112–1113. 272. Guidry JB, Ogbum CL, Griffin EM: Fatal autoimmune
245. Shvidel L, Shtarlid M, Klepfish A, et al: Evans syndrome hemolytic anemia associated with ibuprofen. JAMA 1979;
complicating fludarabine treatment for advanced B-CLL. Br J 242:68–69.
Haematol 1997;99:706. 273. Patmas MA, Willbom SL, Shankel SW: Acute multisystem
246. Sen K, Kalaycio M: Evans syndrome precipitated by fludara- toxicity associated with the use of nonsteroidal antiinflamma-
bine therapy in a case of CLL. Am J Hematol 1999;61:219. tory drugs. Arch Intern Med 1984;144:519–521.
316 Immune Hemolytic Anemias

274. Prescott LF, Illingworth RN, Critchley JAJH, et al: Acute 300. DeTorres OH: Hemolytic anemia and pancytopenia induced
haemolysis and renal failure after nomifensine overdosage. Br by cefoxitin. Drug Intell Clin Pharm 1983;17:816–818.
Med J 1980;281:1392–1393. 301. Duran-Suarez JR, Trujillo J, Prat I, et al: Anti-cephalosporins
275. Woodliff HJ, Wallace PF: Haemolytic anaemia caused by and immune complexes. Transfusion 1982;22:541–542.
nomifensine. Med J Aust 1986;144:163. 302. Salama A, Göttsche B, Schleffer T, et al: “Immune complex”
276. Martlew VJ: Immune haemolytic anaemia and nomifensine mediated intravascular hemolysis due to IgM cephalosporin
treatment in north west England 1984–85: Report of six cases. dependent antibody. Transfusion 1987;27:460–463.
J Clin Pathol 1986;39:1147–1150. 303. Garratty G, Postoway N, Schwellenbach J, et al: A fatal case of
277. Conlon KC, Urba WJ, Smith JW, et al: Exacerbation of ceftriaxone (Rocephin)-induced hemolytic anemia associated
symptoms of autoimmune disease in patients receiving alpha- with intravascular immune hemolysis. Transfusion 1991;31:
interferon therapy. Cancer 1990;65:2237–2242. 176–179.
278. Andriani A, Bibas M, Callea V, et al: Autoimmune hemolytic 304. Ehmann WC: Cephalosporin-induced hemolysis: A case report
anemia during alpha interferon treatment in nine patients and review of the literature. Am J Hematol 1992;40:121–125.
with hematological diseases. Haematologica 1996;81: 305. Chenoweth CE, Judd WJ, Steiner EA, Kauffman CA:
258–260. Cefotetan-induced immune hemolytic anemia. Clin Infect Dis
279. Landau A, Castera L, Buffet C, et al: Acute autoimmune 1992;15:863–865.
hemolytic anemia during interferon-alpha therapy for chronic 306. Wagner BKJ, Heaton AH, Flink JR: Cefotetan disodium-
hepatitis C. Dig Dis Sci 1999;44:1366–1367. induced hemolytic anemia. Ann Pharmacotherapy 1992;26:
280. Stavroyianni N, Stamatopoulos K, Viniou N, et al: 199–200.
Autoimmune hemolytic anemia during alpha-interferon treat- 307. Dhawan M, Kiss JE, DiAddezzio NA, Triulzi DJ: Fatal cefote-
mentin a patient with chronic myelogenous leukemia. Leuk tan-induced immune hemolysis [abstract]. Blood 1993;82:582a.
Res 2001;25:1097–1098. 308. Lo G, Higginbottom P: Ceftriaxone induced hemolytic anemia
281. Akard LP, Hoffman R, Elias L, et al: Alpha-interferon and [abstract]. Transfusion 1993;33:25S.
immune hemolytic anemia. Ann Intern Med 1986;105:306. 309. Eckrich RJ, Fox S, Mallory D: Cefotetan-induced immune
282. Radin AL, Buckley P, Duffy TP: Interferon therapy for agno- hemolytic anemia due to the drug-adsorption mechanism.
genic myeloid metaplasia complicated by immune hemolytic Immunohematology 1994;10:51–54.
anemia. Hematol Pathol 1991;5:83–88. 310. Peano GM, Menardi G, Quaranta L, et al: A rapidly fatal case
283. Fattovich G, Giustina G, Favarato S, et al: A survey of adverse of immune haemolytic anaemia due to cefotetan. Vox Sang
events in 11,241 patients with chronic viral hepatitis treated 1994;66:84–85.
with alfa interferon. J Hepatol 1996;24:38–47. 311. Ogburn JR, Knauss MA, Thapar K, et al: Cefotetan-induced
284. Fornaciari G, Bassi C, Beltrami M, et al: Hemolytic anemia immune hemolytic anemia (IHA) resulting from both drug
secondary to interferon treatment for chronic B hepatitis. J Clin adsorption and immune complex mechanisms [abstract].
Gastroenterol 1991;13:356–357. Transfusion 1994;34:27S.
285. Barbolla L, Paniagua C, Outeirino J, et al: Haemolytic anaemia 312. Mohammed S, Knoll S, vanAmburg A III, Mennes PA:
to the alpha-interferon treatment: A proposed mechanism. Vox Cefotetan-induced hemolytic anemia causing severe hypoph-
Sang 1993;65:156–157. osphatemia. Am J Hematol 1994;46:369–370.
286. Sacchi S, Kantarjian H, O’Brien S, et al: Immune-mediated and 313. Bernini JC, Mustafa MM, Sutor LJ, Buchanan GR: Fatal hemo-
unusual complications during interferon alfa therapy in lysis induced by ceftriaxone in a child with sickle cell anemia.
chronic myelogenous leukemia. J Clin Oncol 1995;13: J Pediatr 1995;126:813–815.
2401–2407. 314. Lascari AD, Amyot K: Fatal hemolysis caused by ceftriaxone.
287. Ballester OF, Spiers ASD, Grattan J: Autoimmune hemolytic J Pediatr 1995;126:816–817.
anemia (AIHA) accopanying treatment with alpha-intereferon 315. Borgna-Pignatti C, Bezzi TM, Reverberi R: Fatal ceftriaxone-
(IFN) in a patient with chronic granulocytic leukemia (CGL). induced hemolysis in a child with acquired immunodeficiency
Blood 1989;74(Suppl.):352. syndrome. Ped Infec Dis J 1995;14:1116–1117.
288. Rotoli B, Formisano S, Alfinito F: Autoimmune haemolytic 316. Scimeca PG, Weinblatt ME, Boxer R: Hemolysis after treatment
anaemia associated with cimetidine. Lancet 1979;2:583. with ceftriaxone. J Pediat 1996;128:163.
289. Rate R, Bonnell M, Chervenak C, et al: Cimetidine and hema- 317. Moallem HJ, Garratty G, Wakeham M, et al: Ceftriaxone-related
tologic effects. Ann Intern Med 1979;91:795. fatal hemolysis in an adolescent with perinatally acquired
290. Donowitz GR, Mandell GL: Beta-lactam antibiotics. N Engl J human immunodeficiency virus infection. J Pediatrics 1998;
Med 1988;318:490–500. 133:279–281.
291. Petri WA: Penicililns, cephalosporins, and other β-lactam antibi- 318. Longo F, Hastier P, Buckley MJM, et al: Acute hepatitis,
otics. In Hardman JG, Limbird LE, Gilman AG (eds): Goodman autoimmune hemolytic anemia, and erythroblastocytopenia
& Gilman’s The Pharmacological Basis of Therapeutics, 10th ed. induced by ceftriaxone. Am J Gastroenterol 1998;93:836–837.
New York: McGraw-Hill, 2001:1189–1218. 319. Garratty G, Leger RM, Arndt PA: Severe immune hemolytic
292. Ferrone S, Zanella A, Scalamogna M: Red cell metabolism in anemia associated with prophylactic use of cefotetan in obstet-
positive direct Coombs test after cephalothin therapy. ric and gynecologic procedures. Am J Obstet Gynecol
Experientia 1971;27:194–195. 1999;9:337–342.
293. York PS, Landes RR, Seay LS: Coombs’ positive reactions asso- 320. Maraspin V, Lotric-Furlan S, Strle F: Ceftriaxone-associated
ciated with cephaloridine therapy. JAMA 1968;206:1086. hemolysis. Wien Klin Wochenschr 1999;111(9):368–370.
294. Fass RJ, Perkins RL, Saslow S: Positive direct Coombs’ tests 321. Meyer O, Hackstein H, Hoppe B, et al: Fatal immune haemol-
associated with cephaloridine therapy. JAMA 1970;213:121–123. ysis due to a degradation product of ceftriaxone. Br J Haematol
295. Schwarz S, Gabl F, Huber H, et al: Positive direct antiglobulin 1999;105:1084–1085.
(Coombs’) test caused by cephalexin administration in 322. Badon SJ, Cable RG: Hemolysis due to cefotetan [abstract].
humans. Vox Sang 1975;29:59–65. Transfusion 1999;39:42S.
296. van Winzum C: Clinical safety and tolerance of cefoxitin 323. Johnson ST, Fueger JT, Gottschall JL: Cefotetan-dependent
sodium: An overview. J Antimicrob Chemother 1978;4(Suppl. antibody cross-reacting with ceftriaxone treated RBCs
B):91–104. [abstract]. Transfusion 1999;39:81S.
297. Meyers BR: Comparative toxicities of third-generation 324. Shammo JM, Calhoun B, Mauer AM, et al: First two cases of
cephalosporins. Am J Med 1985;79(Suppl. 2A):96–102. immune hemolytic anemia associated with ceftizoxime.
298. Forbes CD, Mitchell R, Craig JA, et al: Acute intravascular Transfusion 1999;39:838–844.
haemolysis associated with cephalexin therapy. Postgrad Med 325. Endoh T, Yagihashi A, Sasaki M, Watanabe N: Ceftizoxime-
J 1972;48:186–188. induced hemolysis due to immune complexes: Case report
299. Manoharan A, Kot T: Cephalexin-induced haemolytic and the epitope responsible for immune complex-mediated
anaemia. Med J Aust 1987;147:202. hemolysis. Transfusion 1999;39:306–309.
Drug-Induced Immune Hemolytic Anemia 317

326. Punar M, Özsü H, Eraksoy H, et al: An adult case of fatal Coomb’s tests due to cephalosporins. Postgrad Med J 1970;
hemolysis induced by ceftriaxone. Clin Microbiol Infect 46(Suppl.):107–109.
1999;5:585–586. 349. Mine Y, Nishida M, Goto S, et al: Studies on direct Coombs
327. Viner Y, Hashkes PJ, Yakubova R, et al: Severe hemolysis reaction by cefazolin in vitro. J Antibiotics 1970;23:575–580.
induced by ceftriaxone in a child with sickle cell anemia. 350. Garratty G, Arndt P, Nance S: Nonimmunological adsorption
Pediatr Infect Dis J 2000;19(1):83–85. of proteins onto red cells treated with second and third gener-
328. Stroncek D, Proctor JL, Johnson J: Drug-induced hemolysis: ation cephalosporins [abstract]. Transfusion 1994;34:69S.
cefotetan-dependent hemolytic anemia mimicking an acute 351. Arndt P, Garratty G: Is severe immune hemolytic anemia, fol-
intravascular immune transfusion reaction. Am J Hematol lowing a single dose of cefotetan, associated with the presence
2000;64:67–70. of “naturally-occurring” anti-cefotetan? [abstract] Transfusion
329. Naylor CS, Steele L, Hsi R, Margolin M, Goldfinger D: 2001;41:24S.
Cefotetan-induced hemolysis associated with antibiotic pro- 352. Arndt PA, Leger RM, Garratty G: Reactivity of “last wash”
phylaxis for cesarean delivery. Am J Obst Gynecol elution controls in investigations of cefotetan antibodies
2000;182:1427–1428. [abstract]. Transfusion 1999;39:47S.
330. Moes GS, MacPherson BR: Cefotetan-induced hemolytic 353. Leger RM, Arndt PA, Ciesielski DJ, et al: False-positive eluate
anemia: A case report and review of the literature. Arch Pathol reactivity due to the low-ionic wash solution used with com-
Lab Med 2000;124(a):1344–1346. mercial acid-elution kits. Transfusion 1998;38:565–572.
331. Marques MB, Carr KD, Brumfield CG, Huang ST: A pregnant 354. Hamilton-Miller JMT, Abraham EP: Specificities of haemagglu-
patient with sickle cell disease and cefotetan-induced immune tinating antibodies evoked by members of the cephalosporin C
hemolysis. Lab Med 2000;31(10):541–543. family and benzylpenicillin. Biochem J 1971;123:183–190.
332. Seltsam A, Salama A: Ceftriaxone-induced immune haemoly- 355. Delafuente JC, Panush RS, Caldwell JR: Penicillin and
sis: Two case reports and a concise review of the literature. cephalosporin immunogenicity in man. Ann Allergy
Intensive Care Med 2000;26:1390–1394. 1979;43:337–340.
333. Ray EK, Warkentin TE, O’Hoski PL, Gregor P: Delayed onset 356. Novalbos A, Sastre J, Cuesta J, et al: Lack of allergic cross-
of life-threatening immune hemolysis after perioperative reactivity to cephalosporins among patients allergic to peni-
antimicrobial prophylaxis with cefotetan. Can J Surg cillins. Clin Exp Allergy 2001;31:438–444.
2000;43:461–462. 357. Cerney A, Pichler W: Allergy to antibacterials: the problem
334. Falezza GC, Piccoli PL, Franchini M, Gandini G, Aprili G: with beta-lactams and sulfonamides. Pharmacoepidemiol
Ceftriaxone-induced hemolysis in an adult [letter]. Transfu- Drug Saf 1998;7:S23–S36.
sion 2000;40:1543–1545. 358. Abraham NC, Petz LD, Fudenberg HH: Cephalothin hyper-
335. Malaponte G, Arcidiacono C, Mazzarino C, et al: Cephalosporin- sensitivity associated with anti-cephalothin antibodies. Int Arc
induced hemolytic anemia in a Sicilian child. Hematology Allergy Appl Immunol 1968;34:65–74.
2000;5:327–334. 359. Petz LD: Immunologic cross-reactivity between penicillin and
336. Calhoun BW, Junsanto T, DeTolve Donoghue M, Naureckas E, cephalosporins: A review. J Infect Dis 1978;137(Suppl.):
Baron JM, Baron BW: Ceftizoxime-induced hemolysis second- S74–S79.
ary to combined drug adsorption and immune-complex mech- 360. Arndt PA, Garratty G: Cross-reactivity of cefotetan and ceftri-
anisms. Transfusion 2001;41:893–897. axone antibodies, associated with hemolytic anemia, with
337. Chai L, Pomper GJ, Ross RL, Neal ZM, Champion MH, Snyder other cephalosporins and penicillin. Am J Clin Pathol
EL: Severe hemolytic anemia and liver damage induced by 2002;118:256–262.
prophylactic use of cefotetan [abstract]. Transfusion 2001; 361. Toy PTCY, Chin CA, Reid ME, et al: Factors associated with
41(Suppl. 1):58S. positive direct antiglobulin tests in pretransfusion patients: A
338. Fueger JT, Bell JA, Gottschall JL, Johnson ST: Ceftazidime- case-control study. Vox Sang 1985;49:215–220.
dependent antibody reacting with untreated red cells in the 362. Heddle NM, Kelton JG, Turchyn KL, et al: Hypergam-
presence of drug [abstract]. Transfusion 2001;41(Suppl. 1):104S. maglobulinemia can be associated with a positive direct
339. Citak A, Garratty G, Ücsel R, et al: Ceftriaxone-induced antiglobulin test, a nonreactive eluate, and no evidence of
haemolytic anaemia in a child with no immune deficiency or hemolysis. Transfusion 1988;28:29–33.
haematological disease. J Pediatr Child Health 2002;38: 363. Garratty G: The significance of IgG on the red cell surface.
209–210. Transfus Med Rev 1987;1:47–57.
340. Afenyi-Annan AN, Judd WJ: Cefotetan induced immune- 364. Brown JL: Incomplete labeling of pharmaceuticals: a list of
mediated hemolysis complicated by thrombocytopenia: “inactive” ingredients. N Engl J Med 1983;309:439–441.
alloimmune or thrombotic? [abstract] Transfusion 2002;52:46S. 365. Law IP, Wickman CJ, Harrison BR: Coombs’-positive hemo-
341. Kim S, Song KS, Kim HO, et al: Ceftriaxone induced immune lytic anemia and ibuprofen. Southern Med J 1979;72:707–710.
hemolytic anemia: Detection of drug-dependent antibody by 366. O’Neil MJ, ed. The Merck Index: An Encyclopedia of
ex-vivo antigen in urine. Yonsei Med J 2002;43:391–394. Chemicals, Drugs, and Biologicals. Whitehouse Station, NJ:
342. Arndt P: Practical aspects of investigating drug-induced Merck Research Laboratories, 2001.
immune hemolytic anemia due to cefotetan or ceftriaxone—a 367. Physicians’ desk reference, 57th ed. Oradell, NJ: Medical
case study approach. Immunohematology 2002;18:27–32. Economics, 2003.
343. Viraraghavan R, Chakravarty AG, Soreth J: Cefotetan-induced 368. Curtis BR, McFarland JG, Aster RH: Enhanced detection of
haemolytic anaemia: A review of 85 cases. Adv Drug React antibodies (DDAb) responsible for drug-induced thrombocy-
Toxicol Rev 2002;21:101–107. topenia (DITP) [abstract]. Transfusion 1993;33:77S.
344. Eastlund T, Mulrooney D, Neglia J, et al: Self-limited immune 369. Osbourne SE, Johnson ST, Weitekamp LA, et al: Enhanced
hemolysis in a child after six days of ceftriaxone therapy detection of drug-dependent antibodies reacting with
[abstract]. Transfusion 2002;42:96S. untreated red blood cells in the presence of drug [abstract].
345. Bateman ST, Hu E, Lane C, et al: Antibody to ceftriaxone in Transfusion 1994;34:20S.
HIV pediatric patients and potential implications for RBC 370. Arndt P, Garratty G: Use of albumin solutions for solubilizing
hemolysis [abstract]. Blood 2002;100(11):48b. certain drugs can decrease binding of drugs to RBCs [abstract].
346. Ferrone S, Mercuriali F, Scalamogna M: Cephalothin positive Transfusion 2002;42:105S.
direct Coombs test: Relationship to serum immunoglobulin 371. Wong KY, Boose GM, Issitt CH: Erythromycin-induced
concentration. Experientia 1971;27:193–194. hemolytic anemia. J Pediatr 1981;98:647–649.
347. Garratty G: Laboratory investigation of drug-induced immune 372. Nance SJ, Ladisch W, Williamson TL: Erythromycin-induced
hemolytic anemia and/or positive direct antiglobulin tests. immune hemolytic anemia. Vox Sang 1988;55:233–236.
Washington, DC: American Association of Blood Banks, 373. Johnson HM, Brenner K, Hall HE: The use of a water-soluble
1980:1–30. carbodiimide as a coupling reagent in the passive hemaggluti-
348. Kosakai N, Miyakawa C: Fundamental studies on the positive nation test. J Immunol 1966;97:791–796.
C H A P T E R 9

Unusual Aspects of
Acquired Immune
Hemolytic Anemias

A. Autoimmune Hemolytic Anemia with a


Negative Direct Antiglobulin Test (DAT)

Patients with acquired INCIDENCE OF DAT-NEGATIVE WAIHA


hemolytic anemia whose red
blood cells (RBCs) do not The exact incidence of WAIHA with a negative DAT is
react with antiglobulin serum not known. Evans and Weiser1 described four patients
(AGS) have been reported by with negative DATs who were clinically similar to
numerous investigators. In some instances, the 37 other patients with serologic abnormalities indica-
hemolysis is clearly not antibody-induced; examples tive of AIHA. Dacie2 so noted persistently negative
include hemolytic anemia caused by oxidant drugs DATs among some patients who otherwise appeared
in patients with glucose 6-phosphate dehydrogenase to have WAIHA. Worlledge and Blajchman3 reported
deficiency, mechanical hemolytic anemias, paroxys- 10 (3%) similar cases in their series of 333 cases con-
mal nocturnal hemoglobinuria, and so on. In other sisting of all types of AIHA; in 8 of these cases, sur-
patients, however, extensive evaluation fails to vival of 51Cr-tagged compatible normal RBCs was
reveal a nonimmunologic etiology, and clinical shortened. Chaplin4 reported that 2% to 4% of patients
findings are suggestive of AIHA associated with with AIHA have a negative DAT. Boccardi and col-
warm autoantibodies (WAIHA). That is, these leagues5 reported that 11 of 98 patients (11%) who
patients destroy transfused normal compatible RBCs appeared to have AIHA had negative DATs.
at a rate approximating destruction of their own During a 10-year period during which we studied
RBCs, thus indicating an extrinsic mechanism for 347 patients with definite AIHA (244 were WAIHA),
RBC destruction. In addition, they usually respond we encountered 27 other patients who appeared to
to steroid therapy and/or splenectomy as do have WAIHA but had negative routine DATs.6 This
patients with more typical serologic findings of represented about 10% of the patients with WAIHA
AIHA. For many such patients, evidence supporting and 7% of all the patients with acquired immune
the hypothesis that these are indeed WAIHAs can be hemolytic anemias we encountered during that
obtained by using techniques that are more sensitive period. Sometimes, small amounts of RBC-bound IgG
than standard serologic procedures. could be detected by methods more sensitive than the
319
320 Immune Hemolytic Anemias

antiglobulin test (AGT). Sometimes, the phenomenon


appeared to be associated with the presence of low-
affinity IgG autoantibodies that eluted from the RBCs
during the in vitro washing of the RBCs, and some-

Number of Donors
times the RBC-bound autoantibody was IgA or IgM,
which was not detectable by AGS routinely used for
the DAT.
Negative
RELATIONSHIP OF ANTIBODY DATs

CONCENTRATION TO RATE OF Positive


DATs
HEMOLYSIS

The inability to demonstrate autoantibodies on the Number of IgG molecules/RBC


RBCs of some patients with AIHA seems to sometimes
result from the concentration of antibody being too low FIGURE 9-1. The expected Gaussian distribution of naturally occurring
RBC-bound IgG. Do the positive DATs found in normal donors represent
for detection by the routine DAT. This was first sug- the outer limit of this distribution? (From Garratty G: Effect of cell-
gested by Gilliland and coworkers.7-9 That such small bound proteins on the in vivo survival of circulating blood cells.
concentrations of IgG autoantibody are capable of pro- Gerontology 1991;37:68–94.)
ducing RBC destruction is supported by observations
with alloantibodies. Mollison and Hughes-Jones10
studied the clearance of Rh-positive RBCs by low con- gators reporting on the quantity of IgG on normal
centrations of Rh antibody. They injected anti-Rh(D) RBCs calculate the amount of IgG per RBC (5–90 [mean
into an Rh-negative person and followed the injection of 39] molecules of IgG per RBC) as if it were spread
by 51Cr-tagged Rh(D)-positive RBCs, demonstrating evenly among all the RBCs.17,18 If the IgG present on
that when RBCs were coated with about 10 molecules normal RBCs is an autoantibody to senescent cell
of antibody per RBC, their T50 survival was approxi- antigen (SCA) and appears predominantly on older
mately 100 hours. They also demonstrated that the RBCs (see later in this chapter), then the calculations
lowest concentration of antibody capable of bringing used by Merry and associates17 and Jeje and cowork-
about RBC destruction in vivo was about 1/50th of the ers,18 which yielded similar results of about 30–40 IgG
lowest concentration detectable by the indirect molecules per RBC in the total RBC population, might
antiglobulin test (IAT) in vitro. This finding, however, be incorrect. Their results could also be interpreted to
must not be taken to mean that all antibodies have the mean that the oldest RBCs (i.e., approximately 1% of
same degree of potency with regard to their ability to the total RBC population) might have several thousand
cause RBC destruction in vivo. Indeed, ample data IgG molecules per RBC. This would certainly explain
indicate that the ability of autoantibodies to produce why these older RBCs react efficiently with
RBC destruction varies. Although hemolytic anemia macrophages, whereas it is hard to explain why RBCs
occurs in some patients who have low concentrations with 30–40 IgG molecules would be phagocytosed.
of antibody on their RBCs, other patients have mild or The RBC-bound IgG of most healthy individuals is
no RBC destruction with a strongly positive DAT (see not detectable by the DAT. As mentioned previously,
Chapter 4). Constantoulakis and associates11 demon- this is probably a quantitative effect (Table 9-1 and
strated that the rate of hemolysis depends more on the
characteristics of the individual antibody than on the
antibody concentration. Although wide variations of
antibody concentration per RBC and hemolytic activity TABLE 9-1. THE STRENGTH OF THE ANTIGLOBULIN
are observed among patients, in the individual patient TEST USING RED CELLS COATED WITH VARYING
with AIHA the rate of RBC destruction is generally CONCENTRATIONS OF IgG AS DETERMINED BY THE
related to the concentration of RBC-bound antibody12,13 COMPLEMENT FIXING ANTIBODY CONSUMPTION
(also see Chapter 4). TEST. (A SINGLE ANTI-IgG ANTIGLOBULIN SERUM
If the theory of Gilliland and colleagues7-9 is correct, WAS USED AT OPTIONAL DILUTION.)
we must accept the notion that macrophages can inter-
act with RBCs sensitized with less than 200 IgG mole- RBCs Sensitized In Vitro with Anti-RhD)
cules per RBC. Not only do they have to interact, but
Antiglobulin IgG Molecules
the interaction must be efficient enough to cause a Test per RBC
hemolytic anemia. If senescent RBCs are removed by
an immune mechanism, there must be a delicate Negative <25–120
balance between the amount of IgG causing normal 1/ + 120
2
daily RBC destruction and the amount causing DAT- 1+ 200
2+ 300–500
negative AIHA (see Fig. 9-1).14-16 IgG is thought to be 3+, 4+ >500
present on the RBCs of most individuals. Most investi-
Unusual Aspects of Acquired Immune Hemolytic Anemias 321

Fig. 9-1). Even if the older RBCs have amounts of IgG are important variables. Gilliland and coworkers7-9
(i.e., >120 molecules/RBC) that should be detectable reported that the RBCs of some patients with 150 mol-
by the AGT, they would be too minor a population to ecules of IgG per cell were agglutinated by AGS, while
form agglutinates readily visible under the conditions RBCs from other patients were not agglutinated even
by which the routine AGT is performed. We do not though they were sensitized with 700 molecules per
know whether the RBC-bound IgG detectable by the RBC. Petz and Garratty6 reported similar results.
DAT in approximately one in 1400 seemingly healthy Dupuy and colleagues21 observed weak agglutination
blood donors represents the upper end of the quanti- of RBCs coated with 100 molecules of anti-Rh(D) per
tative range typical of the normal gaussian distribu- RBC by some AGS; with other AGS, however, 10–20
tion of RBC-bound IgG (Fig. 9-1), or whether it times more RBC-bound antibody was required for
suggests autoantibodies to SCA or a different popula- agglutination. Other observers, using RBCs coated
tion of IgG molecules.14-16 We do not know whether with either IgG anti-A or anti-Rh(D), have observed
there is any relationship between the pathogenicity of minimally detectable agglutination by AGS, with anti-
certain IgG autoantibodies and increased amounts of body concentrations ranging from 100 to 500 mole-
RBC-bound SCA autoantibody, or whether pathoge- cules per RBC.22,23
nic autoantibodies are always of a different specificity. Results carried out in our laboratory with RBCs sen-
There is some evidence that SCA autoantibody can be sitized in vitro with anti-Rh(D) are shown in Table 9-1.
pathogenic at times (see Chapter 7). Negative AGT results were obtained with RBCs coated
The situation can be even more complicated than with fewer than 120 molecules per RBC. Progressively
that described in the foregoing discussion. Masouredis stronger reactions occurred with higher concentrations
and colleagues19 showed that the IgG eluted from the of IgG. The concentration of IgG on the RBCs was
RBCs of DAT-positive, seemingly healthy blood donors determined using the complement-fixing antibody
was composed of at least two antibody populations. consumption test described later in this chapter.
The first was an IgG autoantibody directed at RBC anti-
gens; the second was an IgG autoanti-idiotype, directed
against the RBC-bound IgG autoantibody (Fig. 9-2).
MEASUREMENT OF SMALL AMOUNTS
The same group also showed that the RBC-bound IgG OF RBC-BOUND IgG
anti-idiotype did not appear to be present in eluates
prepared from the RBCs of DAT-positive patients with Small amounts of IgG RBC-bound autoantibodies can
AIHA.20 Masouredis and associates19,20 suggested that be measured by techniques not available in most
the IgG autoanti-idiotype could play a role in protect- laboratories. These include the complement fixation
ing the IgG-sensitized RBCs in normal donors from antibody consumption (CFAC) test, the radiolabeled
being destroyed at an abnormal rate, thus preventing anti-IgG test, the enzyme-linked antiglobulin test
AIHA in DAT-positive donors. (ELAT), and flow cytometry.6-9,24-47
Gilliland and coworkers8 reported that the RBCs of
five of six patients with acquired hemolytic anemia and
SENSITIVITY OF THE DAT a persistently negative DAT were found to have abnor-
mal quantities of IgG as measured by the CFAC test.
The concentration of IgG antibody on the RBCs that is The amount of IgG per RBC ranged from 70 to 434 mol-
required for a positive DAT is quite variable. The char- ecules, compared with 35 or fewer molecules of IgG per
acteristics of the RBCs, the RBC antibody, and the AGS RBC found on the RBCs of normal persons. RBCs of
patients with other types of hemolytic anemia had
fewer than 35 molecules per RBC. These included RBCs
Untreated RBC IgG(Rh) - Coated RBC of two patients with chronic hemolytic anemia due to
Agglutination* IAT Agglutination*
cold agglutinins, two patients with paroxysmal noctur-
O + O nal hemoglobinuria (PNH), two with hereditary sphe-
Ab1
rocytosis, and one with burr cells related to chronic
R liver disease. In patients who had abnormal concentra-
B
C O O + tions of IgG on their RBCs, eluates prepared from
Ab2 50–200 mL of blood and concentrated to a final volume
of 1 mL reacted in the IAT. Eluates prepared from RBCs
of normal subjects or from subjects with other types of
FIGURE 9-2. Eluates from RBCs of DAT-positive blood donors contained
hemolytic anemia were nonreactive. Further evidence
two antibody populations: Ab1 and Ab2. Ab1 was the RBC auto- that the IgG in the RBC eluates had antibody character-
antibody that reacted with untreated RBCs by IAT but did not aggluti- istics was shown by their specificities. Three of the
nate them or lgG(Rh)-coated RBCs. Ab2 was an IgG autoantibody to an eluates reacted with RBCs of common Rh phenotypes
epitope on the lgG RBC autoantibody (said to be anti-idiotype). Ab2 did but did not react with Rhnull RBCs, thus reflecting the
not react with untreated RBCs but directly aggultinated lg(Rh)-coated
RBCs. (*Agglutination= agglutination at 37ºC.) (From Garratty G: diversity of specificity seen in patients with AIHA
Autoimmune hemolytic anemia. In Garratty G (ed): Immunolobiology of and positive DATs (see Chapter 7). One of the six
Transfusion Medicine. New York: Dekker, 1994:493–521.) patients with acquired hemolytic anemia had a normal
322 Immune Hemolytic Anemias

concentration of IgG on her RBCs as measured by the sensitization of the patients’ RBCs in 11 (41%) instances,
CFAC test, and no IgG, IgA, or IgM RBC antibodies using an anti-C3 containing a potent anti-C3d that was
could be identified in a concentrated eluate prepared prepared in our laboratory. In most of these instances,
from 200 mL of blood. Further, no free antibody was the antiglobulin test was only weakly positive. Also, in
detected in her serum; however, survival of transfused one instance, we obtained a weakly positive reaction
normal compatible RBCs was greatly shortened. using an anti-IgG made in our laboratory.
Gilliland9 extended these findings by reporting on an Autoantibodies were detected in the serum or
additional nine patients. The amount of IgG per RBC in eluate in 11 of 27 patients (41%). For some patients,
these patients ranged from 76 to 350 molecules. Six of eluates prepared from several mL of RBCs and con-
the nine responded to corticosteroid therapy, one did centrated two to five times resulted in a positive reac-
not improve, one died of chronic lymphocytic leukemia tion. Most often, the autoantibody was detected in the
before results of therapy could be assessed, and one serum using enzyme-treated RBCs.
patient was lost to follow-up. The patient who showed The CFAC test revealed more than 25 molecules of
no improvement had the same quantity of IgG on his IgG per RBC in all patients and between 100 and
RBCs three weeks later. Follow-up quantitative RBC 300 molecules of IgG per RBC in 15 of the 27 cases
antibody data were obtained for some of the patients (56%).6 In five instances, more than 400 molecules of
who responded to therapy, and these data revealed a IgG per RBC were detected; this result includes one
decrease in the amount of antibody per RBC. extraordinary case of a patient with recurrent
We performed 82 CFAC tests on 59 subjects who episodes of brisk hemolysis (see Patient 1), in whom
were healthy or who had disorders other than idio- 1400 molecules of IgG per RBC were found in spite of
pathic acquired hemolytic anemia.6 In addition, we a negative DAT.
studied 27 patients who were referred to our labora- Information concerning response to therapy was
tory with a diagnosis of acquired hemolytic anemia available for 17 patients, and follow-up CFAC tests
with a negative DAT.6 Results in the former group of were performed for seven of these.6 Six of these
patients are summarized in Table 9-2. Of 54 studies on patients revealed marked clinical improvement in
31 normal subjects, only one was positive—that is, response to steroids or splenectomy. The subsequent
only one indicated more than 25 molecules of IgG per CFAC test revealed fewer than 25 molecules of IgG per
RBC. Of 11 patients with hereditary spherocytosis, RBC for five patients, but in one patient, 228 molecules
one CFAC test was positive in a patient who also had of IgG were present after therapy (compared with 173
Hodgkin’s disease. Tests in one patient with pyruvate prior to therapy) in spite of clinical improvement. One
kinase deficiency and two with Gilbert syndrome patient who did not improve as a result of treatment
with mild reticulocytosis were negative, as were tests with steroids and splenectomy had 180 molecules of
for three patients who had had splenectomy for a IgG per RBC before therapy and 607 when retested 2
myeloproliferative disorder. Only one of 11 patients years later. Twenty-two of the patients had no associ-
with mild hemolysis associated with the presence of a ated disorder and were diagnosed as having idiopathic
prosthetic heart valve had a positive CFAC test. AIHA, although one of these had a history of idiopathic
Our experience with 27 patients who were referred to thrombocytopenic purpura. Five patients had active
our laboratory with a diagnosis of acquired hemolytic associated disease. Spherocytes were noted on the
anemia with a negative DAT are summarized in Table peripheral blood film of seven patients, although this
9-3.6 Although the DAT was negative in all cases in the information is not indicated in the table. This finding
referral laboratory, we were able to detect complement suggests that the interaction of antibody-coated RBCs
with the cells of the reticuloendothelial system (see
Chapter 4) occurred in spite of the small numbers of
TABLE 9-2. COMPLEMENT FIXING ANTIBODY IgG molecules present on the RBCs.
CONSUMPTION TEST IN NORMAL SUBJECTS While accumulating this experience, we also obser-
AND IN PATIENTS WITH DISORDERS OTHER ved five patients who were referred with a diagnosis of
THAN IDIOPATHIC ACQUIRED HEMOLYTIC ANEMIA idiopathic acquired hemolytic anemia despite a nega-
tive DAT and in whom the CFAC test indicated that the
Number with patients’ RBCs were coated with fewer than 25 mole-
Number of >25 Molecules cules of IgG per RBC (the lower limit of sensitivity of the
Diagnosis Studies of IgG/RBC test in our laboratory).6 In four of these patients, C3d
was detectable on the patients’ cells by the DAT, but in
Normal persons 54 1
Hereditary spherocytosis 11 1*
none were autoantibodies detectable in the serum or
Pyruvate kinase deficiency 1 0 eluate. Whether these patients’ hemolytic anemia was
Gilbert syndrome 2 0 also immunologically mediated is conjectural.
Postsplenectomy 3 0 The following case reports illustrate the value of
Mechanical hemolytic anemia 11 1 sensitive serologic tests and the CFAC test in evaluat-
(prosthetic heart valves)
ing patients with acquired hemolytic anemia with a
* Patient also had Hodgkin’s disease. negative DAT.
Unusual Aspects of Acquired Immune Hemolytic Anemias 323

TABLE 9-3. WARM ANTIBODY AUTOIMMUNE HEMOLYTIC ANEMIC WITH A NEGATIVE DIRECT
ANTIGLOBULIN TEST

Test for Autoantibody


Serum Eluate CFAC Test:
DAT in DAT in Authors’ Enzyme IgG Molecules
Referral Laboratory Treated per RBC Associated
Laboratory IgG C3d IAT RBC IAT (Normal ≤ 25) Therapy Course Disease

1 Neg Neg 11⁄2+ Neg Neg Neg 144 None Improved


2 Neg Neg 2+ Neg Neg Neg >400 History of ITP
3 Neg Neg Neg Neg Neg NT 126 Wiskott-Aldrich
4 Neg Neg 1+ Neg 31⁄2+ Neg 251 Steroids Improved
5 Neg Neg Neg Neg Neg Neg 93
6 Neg Neg Neg Neg Neg NT 59 Steroids Improved
7 Neg Neg 1+ Neg 11⁄2+ Neg 234
8 Neg 1⁄ + 1⁄ + 1⁄ + 31⁄2+ Neg 126
2 2 2
9 Neg Neg Neg Neg Neg Neg >400 Steroids Improved
10 Neg Neg Neg Neg 1⁄ + Neg 166
2
11 Neg Neg 1⁄ + 2+ 31⁄2+ 1⁄ + 173 Steroids Improved
2 2
12 Neg Neg 1+ Neg Neg NT 339
13 Neg Neg Neg Neg 3+ Neg 243 Steroids Persistent Chronic persistent
hemolysis: hepatitis
expired
14 Neg Neg Neg Neg Neg Neg 54 Steroids Unchanged Metastatic
carcinoma:
microangiopathic
hemolytic
anemia
15 Neg Neg Neg 1+ Neg NT 128 Steroids Improved
16 Neg Neg Neg Neg Neg 2+ 185 Steroids Improved
17 Neg Neg Neg Neg Neg Neg 70 Steroids Unchanged TTP
18 Neg Neg Neg Neg Neg NT >400 Steroids Improved
19 Neg Neg Neg Neg Neg 1⁄ + 95 Steroids Improved
2
20 Neg Neg 1+ Neg Neg NT 122 Carcinoma of
ovary
21 Neg Neg Neg Neg Neg Neg 127
22 Neg Neg Neg 1⁄ + Neg Neg 290 Steroids Improved
2
23 Neg Neg Neg NT NT NT 59
24 Neg Neg 3+ Neg 2+ 1+ 1400 Splenectomy Long term
remission
25 Neg Neg 21⁄2+ Neg Lysis 11⁄2+ 138 Splenic irradiation Marked temporary
improvement
26 Neg Neg 1⁄ + Neg Neg Neg 180 Prednisone and Unchanged
2
splenectomy
27 Neg Neg Neg Neg Neg Neg 640 Steroids Rapid improvement

Neg = Negative
NT = Not tested
ITP = Idiopatnic thrombocytopenic purpura
TTP = Thrombotic thrombocytopenic purpura

P A T I E N T 1 : A 12-year history of episodic malaise, pallor, and (by the fourth or fifth day) jaundice and
acquired hemolytic anemia with a negative symptoms of anemia. The nadir of hemoglobin depression
DAT.6 A 49-year-old male had a 12-year history of recur- was generally in the range of 5–7 g/dL and occurred
rent episodes of hemolytic anemia. He developed malaise, 7–10 days after the onset of symptoms. Reticulocytopenia
fatigue, gastrointestinal symptoms, jaundice, and dark (1% or less) was present during the period of progressive fall
urine. An acute hemolytic anemia was diagnosed, and in hemoglobin, usually continuing for about 72 hours after
there was no evidence of precipitating events. The hemoly- reaching the nadir. Leukopenia and thrombocytopenia also
sis resolved spontaneously without therapy, and over the occurred, with the white cell count dropping to about
next 2 years, the patient had recurrent episodes at approx- 2000/μL with a normal differential and the platelets falling
imately 60–90 day intervals. The episodes gradually to about 80,000/μL. Then followed a progressive reticulo-
decreased in severity, and 2 years later, he was free of cytosis to a value of about 20% over the succeeding 7–10
symptoms. Six years later, he developed recurrent attacks days and complete recovery at the end of approximately 3
consisting of progressive weakness, darkening of the urine, weeks. At that time, the patient’s hemoglobin was in the
324 Immune Hemolytic Anemias

range of 14 g/dL, and the reticulocyte count, white cell When referred for immunohematologic evaluation
count, and platelet count were normal. These episodes con- 1 year later, his DAT was negative with anti-IgG but mod-
tinued to recur at 60–90 day intervals for the next 4 years, erately strongly positive with anti-C. His serum contained
during which time numerous studies were undertaken. an antibody that reacted equally with all enzyme-treated
The patient was questioned repeatedly about any envi- RBCs of a panel. An alloantibody (anti-Wra) was also
ronmental factor relating to occupation or hobbies that detectable by IAT. An eluate prepared from the patient’s
might be of significance in precipitating attacks, but none RBCs contained a weak antibody without evident
could be identified. There was no history of drug ingestion. specificity. In spite of the negative DAT using anti-IgG, the
Physical examination was remarkable for the presence of CFAC test revealed a surprising value of 1400 molecules
mild tachycardia and icterus during episodes of marked of IgG per RBC at a time when the patient’s hemoglobin
anemia. In addition, he had persistent splenomegaly, the was 5.9 g/dL. A repeat test in January 1979, when the
edge of the spleen usually palpable 4 cm below the left hemoglobin was 13.6 g/dL, revealed 396 molecules of
costal margin. IgG per RBC.
Laboratory data revealed characteristic findings of A splenectomy was performed. The spleen weighed
hemolysis during periods of progressive anemia. In a 1164 g and revealed congestion of the red pulp, follicu-
typical episode, the total bilirubin was 1.6 mg/dL with lar center hyperplasia, and abundant iron. Subsequently
0.3 mg direct reacting, haptoglobin absent, LDH 90, and (13 years after his first hemolytic episode), the patient was
urine hemosiderin weakly positive. The RBC morphology asymptomatic and had no further hemolytic episodes
was essentially normal, although an occasional target cell during a 3-year follow-up period. His hemoglobin was
and rare spherocytes were present. A bone marrow aspi- 17.2 g/dL, hematocrit was 51.4%, white cell count
rate performed at a point of maximum anemia revealed was 11,200/μL, platelet count was 329,000/μL, and
intense erythroid hyperplasia with some megaloblastoid reticulocytes were 1.7%. A repeat CFAC test performed
features. A repeat bone marrow study performed during a 6 months after splenectomy revealed fewer than 25 mol-
phase of remission revealed some residual erythroid ecules of IgG per RBC. Three years later, the CFAC test
hyperplasia that was normoblastic. An RBC survival study revealed 240 molecules of IgG per RBC; his DAT was
using 51Cr-labeled RBCs was begun near the onset of an negative with anti-IgG and anti-C3, and no antibody was
episode of hemolysis. The curve initially began with a detected in his serum or in a RBC eluate.
steady exponential fall revealing a halftime of 17 days Comment: This patient had a long history of recurrent
(normal = 25 to 35 days). Suddenly, on the 11th day of attacks of hemolysis without evident cause. Attacks
the study, the chromium disappearance became markedly occurred every 60–90 days, and his hemoglobin fell to
accelerated with a halftime of 3 days. Excessive splenic as low as 3.5 g/dL. Immunohematologic evaluation
sequestration was noted. Another RBC survival study was (including the use of the CFAC test) revealed evidence of
begun when the patient’s hematocrit reached 12.5%. WAIHA. The patient responded to splenectomy with com-
During the study, his hematocrit level rose to 24% on the plete cessation of episodes of hemolysis, and follow-up
10th day and to 38% on the 17th day. The disappear- immunohematologic evaluations revealed significant
ance curve of the radioactive chromium was within improvement.
normal limits.
Extensive diagnostic studies to determine the cause of
the hemolysis were unproductive.Tests performed included P A T I E N T 2 : Acquired hemolytic anemia with
the DAT, Ham’s acid serum test, the sucrose hemolysis test, a negative DAT with anti-E, occurring in the
osmotic fragility before and after incubation at 37°C for RBC eluate of an E-negative patient.44 A previ-
24 hours, hemoglobin electrophoresis, quantitation of ously untransfused 20-year-old male presented with a 7-
hemoglobins A and E, isopropanol and heat tests for day history of malaise, fatigue, jaundice, dark urine, and
unstable hemoglobins, and biochemical measurement splenomegaly. Hemolytic anemia was indicated by a
(RBC G-6PD, 6-phosphogluconic hydrogenase, hexoki- hemoglobin of 8.7 g/dL, reticulocyte count 8%, lactic
nase, pvruvate kinase, aldolase, catalase, pyruvate, and dehydrogenase 389 IU/L, bilirubin 4.3 mg/dL (direct
lactate). 0.1 mg/dL), and undetectable serum haptoglobin. Tests
Prednisone at a dose of 80 mg per day was given at the for nonimmunologic causes of hemolytic anemia were
onset of symptoms. This regimen had no significant effect on negative. The DAT was repeatedly negative with poly-
the progression of the hemolytic episode. The patient was specific, anti-IgG, -IgA, and -IgM antisera. The patient’s
also treated with folic acid without effect. Transfusions were serum contained a weak anti-I, and anti-E strongly reac-
given during his first episode of hemolysis, but thereafter the tive as determined by IAT, and an antibody reactive
patient learned to tolerate symptoms in spite of striking drops against all RBCs tested. The latter antibody reacted
in hemoglobin. When his disease became progressively weakly by the IAT but strongly against enzyme-treated
more severe 12 years later, however, he was given 2 units RBCs (titer 160). Eluates from the patient’s RBCs reacted
of RBCs during each of two successive hemolytic episodes only with E+ RBCs. The patient was E-negative. He was
after his hemoglobin had dropped to 3.5 g/dL. treated for WAIHA with high doses of prednisone. By the
Unusual Aspects of Acquired Immune Hemolytic Anemias 325

12th day, his response allowed the medication to be P A T I E N T 4 : Relapsing AIHA in pregnancy in
tapered, and by 1 month from the onset of treatment, lab- a patient with a negative DAT.46 This interesting
oratory studies had returned to normal. The DAT case is discussed in detail in the section on AIHA During
remained negative; following recovery, however, anti-E Pregnancy on page 348.
could not be eluted from the RBCs. Anti-E remained in his
serum, and the titer of the enzyme-reactive antibody had
decreased to 16. It was suggested that the anti-E–like
antibody represented autoanti-Hr preferentially reacting
The CFAC Test in Patients with Sickle
with E+ RBCs (see Chapter 7). Cell Disease (SCD) and Other
Our experience has indicated that when IgG antibody Hemoglobinopathies
is present in an eluate prepared from RBCs, the CFAC test In extending our studies to include a representative
is always abnormal. Thus, we performed the assay only sample of other nonimmune hemolytic anemias, we
on RBCs obtained 5 days following remission, at which performed CFAC tests on RBCs from patients with a
time the eluate was nonreactive. The CFAC revealed variety of congenital hemolytic anemias.6,47 Table 9-4
normal findings (<25 molecules IgG/RBC). shows the results. Rather surprisingly, we found that
Ganly and associates45 described a similar case with 39 of 62 patients (63%) who were homozygous for
an anti-Jka eluted from a Jk(a–) DAT-negative patient with hemoglobin S had a mean of 195 (median of 290) and
Evans’s syndrome. Later, the patient’s DAT became posi- a maximum of 890 molecules of IgG per RBC; these
tive, and the patient’s RBCs at that time were found to be patients would have fit our criteria for DAT-negative
Jk(a+). The specificities in these two cases are similar to WAIHA.
the mimicking antibodies discussed in Chapter 7. The patients ranged in age from 11 months to
30 years, the hemoglobin from 6.0 to 11.1 g/dL, and
the reticulocytes from 0.8% to 20%. We observed no
P A T I E N T 3 : WAIHA erroneously diagnosed significant correlations between the number of IgG
as cold agglutinin syndrome. A 17-year-old molecules on patients’ RBCs and the severity of their
male was admitted to the hospital for evaluation of anemias, the incidence of painful sickle cell crisis, the
severe hemolytic anemia. Physical examination revealed reticulocyte counts, or blood transfusion histories.
an icteric young man in no acute distress. The spleen RBC survival studies were not performed.
was palpable 2 cm below the left costal margin. The Eluates were made from the RBCs of 23 of the 29
hemoglobin was 7.6 g/dL, reticulocytes 10.8%, white patients with SCD who had increased numbers of IgG
cell count 7600/μL, serum bilirubin 7.2 mg/dL (0.8 molecules per RBC. Although only small volumes of
direct reacting), urine hemoglobin 4+, and the mono RBCs were generally available for preparing eluates,
spot test negative. The peripheral blood film revealed six of the 23 eluates (26%) demonstrated antibody
anisocytosis, polychromasia, and numerous sphero- activity against normal RBCs by the IAT. In each case,
cytes. The patient required transfusion of 10 units of
blood in a 10-day period to maintain a hematocrit level
of about 25%. The DAT was negative. The only free
antibody detectable in his serum was a cold agglutinin TABLE 9-4. RESULTS OF CFAC TESTS ON NORMAL
with anti-I specificity that had a maximum titer of 512 at PERSONS AND PATIENTS
4°C and demonstrated weak reactivity up to 22°C. The
Donath-Landsteiner test was negative. A diagnosis of Number of Number of
atypical cold agglutinin syndrome was made, and the Subjects Patients with
Diagnosis Tested Positive Results*
patient was treated by keeping the temperature of his
room at 37°C. Brisk hemolysis continued, however. He Normal persons 42 1
received 16 additional units of RBCs during the next Hemolytic anemias
month without his hematocrit level rising above 30%. Hereditary spherocytosis 11 1
Studies on blood referred to our laboratory revealed Pyruvate kinase deficiency 1 0
Mechanical hemolytic anemia
the DAT was negative using anti-IgG, -IgA, -IgM, and
(prosthetic heart valves) 11 1
anti-C3. No warm autoantibodies were detected in the Hemoglobin abnormalities
serum by IAT or by agglutination of enzyme-treated Sickle cell anemia (S–S) 62 39
RBCs. A cold agglutinin was present to a titer of 8 at Sickle cell disease (S–C,
4°C in saline; it did not cause agglutination in saline or S–D, S/β-thalassemia) 14 8
Sickle cell trait (A-S) 57 7
albumin at 25°C. The CFAC test revealed more than β-thalassemia major 6 5
400 molecules of IgG per RBC. The patient was subse-
quently treated with therapy appropriate for WAIHA and * A positive result indicates >25 molecules of lgG/red cell.
From Petz LD, Yam P, Wilkinson L, Garratty G, Lubin B, Mentzer W: Increased
had a partial response to steroids, but he subsequently IgG molecules bound to the surface of red blood cells of patients with sickle
required splenectomy before achieving remission. cells anemia. Blood 1984;64:301–304.
326 Immune Hemolytic Anemias

specificity tests revealed no definite specificity, thus detail in the section later in this chapter regarding the
suggesting autoantibody activity. development of autoantibodies and autoimmune
One patient’s case history (Patient 5) illustrates the hemolytic anemia following transfusion. In addition,
possible significance of these studies. Constantoulakis and associates11 suggested that above
a certain level (“hemolytic threshold”), minute in-
P A T I E N T 5 : The patient was a 24-year-old female creases of RBC-bound antibody induce pronounced
with SCD who had received multiple transfusions during effects on RBC survival. These authors emphasized that
childhood, although in the past 4 years she had received an acute, explosive hemolytic crisis in a patient with an
only 3 units of RBCs, which had been administered otherwise chronic, compensated AIHA could result
6 months previously. Her hematocrit level was 18%, and from a relatively small increment in antibody produc-
her reticulocyte count was 18.1%. She was admitted to tion, and that this increase in antibody production
the hospital for an elective cholecystectomy, and the deci- would not be detected by conventional serologic tests.
sion was made to transfuse her until her hematocrit level The patient’s hyporegenerative crisis could also be
was about 36% in an attempt to reduce her anesthetic immunologically mediated, as there is ample precedent
risk. Her DAT was negative, but a CFAC test done at the for reticulocytopenia as part of the presentation for an
onset of the transfusions revealed 533 molecules of IgG acute AIHA (see Chapter 3, page 69).
per RBC, and a RBC eluate was weakly reactive with no In addition to 98 SCD patients, we used the CFAC
definable specificity. She was known to have had allo- assay to test RBCs from 59 patients with sickle cell
anti-M and anti-C in the past, as well as multiple HLA anti- trait, 11 patients with hemoglobin S-C, nine patients
bodies, and she was therefore transfused with with thalassemia syndrome (including heterozygous
leukocyte-poor M–, C–RBCs, even though anti-M and and homozygous thalassemia), and patients doubly
anti-C were not detectable at this time. heterozygous for sickle cell hemoglobin and tha-
After transfusion of 8 units of leukocyte-poor RBCs lassemia.47 Ten patients with sickle cell trait (17%)
over a 4-day period, her hematocrit level reached yielded abnormal results. Six patients had more than
36.8%. Two days later, she began to have shaking 250 molecules of IgG per RBC on at least one occasion.
chills, abdominal pain, and fever, and she experienced Of the five patients in whom repeat samples were
a life-threatening hemolytic reaction, with her hematocrit obtained, only one patient was found to have consis-
level dropping to 13% in 48 hours and ultimately reach- tently abnormal results. Six of the 11 patients with
ing a low of 7.4%. The serum hemoglobin reached 283 hemoglobin S-C had increased numbers of IgG mole-
mg/dL, and she had hemoglobinuria and hemosiderin- cules per RBC, as did six of the nine patients with tha-
uria. She also suffered a hyporegenerative crisis, with a lassemia syndromes.
reticulocyte count of 0. There are some interesting findings related to the IgG
A DAT was negative. Repeat screening of her serum for on normal RBCs and RBCs of patients with SCD. Kay48
alloantibodies revealed negative results except for very has suggested that as RBCs age, they develop an SCA,
weak reactions with enzyme-treated cells, with no anti- and such RBCs can take up IgG autoantibody, leading
body specificity defined. She was treated with high doses to removal of the older RBCs by macrophages in the
of corticosteroids and two additional units of leukocyte- RES (see associated literature reviewed by Garratty).14
poor packed RBCs, and she gradually improved to a Green and colleagues49 confirmed other researchers’
hematocrit level of 19.8%. findings that the amount of IgG was greater on old than
This patient’s course was consistent with that of a on younger RBCs when RBCs are separated by density.
delayed transfusion reaction, although there were no They also reported similar findings when SCD patients
detectable alloantibodies. Our data offer a possible were studied; older sickle cells contained two to three
alternative explanation in that autoimmunization might times more RBC-bound IgG than younger sickle cells.
have played a key role. Green and associates50 went on to determine that irre-
Autoimmunization was suggested by the presence of versibly sickled cells (ISC) formed in vivo alterations in
533 molecules of IgG on the patient’s RBCs at the onset the topography of the membrane proteins on the exter-
of transfusions, by an RBC eluate that was weakly reac- nal RBC surface that would cause preferential in vivo
tive by the IAT, and by the response to corticosteroids. It is binding of autologous IgG. Using flow cytometry, they
possible that the transfusions stimulated the further produc- demonstrated a nonuniform distribution of RBCs
tion of autoantibodies, which also reacted with the donor within each density-separated RBC fraction exhibiting
RBCs to cause a transient AIHA. The lack of strongly reac- fluorescence as a result of labeling with FITC-
tive serologic abnormalities usually found in AIHA could conjugated anti-IgG. Each RBC fraction contained at
be analogous to the cases of AIHA with a negative DAT least three distinct subpopulations:
described previously.
1. A weakly fluorescent subpopulation, representing
The suggestion that an augmentation of autoanti- most of the RBCs in the dense (older) population,
body production could be of significance in this clinical and considered to be IgG negative
setting is strengthened by reports of numerous investi- 2. A comparatively small subpopulation represent-
gators concerning the production of autoantibodies ing 1% to 12% of total RBCs, that were coated with
after immunization with RBCs. This is reviewed in IgG
Unusual Aspects of Acquired Immune Hemolytic Anemias 327

3. A small (<1%) subpopulation that showed very studies using various carbohydrates and thermal strip-
bright fluorescence, indicating comparatively ping indicated that at least a proportion of the IgG mol-
larger amounts of RBC-bound IgG ecules found on the thalassemic RBCs were specifically
reactive, with terminal galactosyl residues on the RBC
The researchers proposed the following role for membrane. IgG antibodies with similar reactivity were
RBC-bound IgG in the transformation of the reversible also demonstrated in normal human serum. These IgG
sickle cell (RSC) to ISC: Immunoglobulin molecules natural antigalactosyl antibodies in normal sera could
bind rapidly to high-affinity membrane proteins that bind to IgG-depleted thalassemic RBCs. Thalassemic
emerge sequentially as the cell undergoes successive RBCs and normal senescent RBCs had been described
sickling-desickling cycles in vivo; IgG binding cross- previously as having reduced amounts of membrane
links and inhibits the regression of the newly emerged sialic acid. Galili and colleagues53 suggested that the
surface proteins into the lipid layer. If the IgG binding IgG galactosyl antibodies interact with newly exposed
proteins are also transmembrane proteins attached to galactosyl residues underlying the sialic acid residues,
the membrane cytoskeleton, cross-linking by surface- and that such interaction could lead to the shortened
bound IgG (even 200–300 molecules per RBC) could life span of thalassemic RBCs and result in sequestra-
inhibit the return of the membrane skeleton to the tion of normal RBCs by the RES.
discoid shape (e.g., after a critical number of sickling- The foregoing findings suggest that there could be
desickling cycles), producing RBCs fixed in the ISC an immune component of the increased RBC destruc-
morphology. The researchers also suggested that it tion in SCD and possibly also in thalassemia.
was possible that at least part of the RBC-bound IgG
(measured in this study for the most dense ISC-
enriched sickle RBC fractions) was the senescent RBC Could Positive Sensitive Assays for
autoantibody, as demonstrated for normal RBCs.48 RBC-Bound IgG (e.g., ELAT Results) Be
Because investigations have shown that sickle RBCs Due to Nonspecific Uptake of IgG Rather
must be chronologically younger (life span in vivo, Than Autoantibody?
10–40 days compared with normal RBCs [life span
~120 days]), their findings suggest the possibility that Toy and coworkers54 and Heddle and associates55
transformations of sickle RBCs to the ISC morphology found that RBCs with positive DATs yielding nonreac-
could be associated with rapid RBC aging in vivo.50 tive eluates commonly were associated with levels of
Hebbel and Miller51 showed that RBCs from SCD gamma globulin at the higher end of the normal range
patients were excessively susceptible to phagocytosis. or hypergammaglobulinemia; this suggested that IgG
They reported that SCD RBCs were 1.03–6.85 times was bound nonspecifically to the RBC membrane.
more adherent to macrophages than normal RBCs Thus, it would seem essential that any interpretation of
and that the degree of adherence correlated signifi- assays for RBC-bound IgG in suspected DAT-negative
cantly with irreversibly sickled cell counts and hema- AIHA be related to serum globulin levels. Garratty and
tologic values reflecting hemolysis rate. SCD RBCs Arndt56 measured the serum IgG levels in 48 patients
spontaneously generate twice the normal amounts of who were thought to have DAT-negative AIHA and
dialdehyde byproducts of lipid peroxidation (i.e., had positive sensitive test results (i.e., a positive ELAT,
malondialdehyde [MDA]), and MDA treatment of a positive direct Polybrene test, and/or a reactive con-
normal RBCs significantly enhances their phagocyto- centrated eluate). Results were compared with a
sis. Hebbel and Miller51 suggested the possibility that control group of 52 patients with a similar suspected
appearance of the SCA on old normal RBCs repre- diagnosis but yielding negative ELAT, Polybrene, and
sents modification of the membrane by MDA. This eluate results. The serum IgG levels were divided into
report prompted Solanki52 to look for evidence of in four arbitrary groups of 0–600, 601–1200, 1201–1800,
vivo erythrophagocytosis (Ep) in SCD patients. He and greater than 1801 mg/dL. Within the first three
examined blood smears of 27 patients and found 10 groups (0–1800 mg/dL), there was no correlation
(37%) to show Ep (i.e., 1 to 6/1000 white blood cells between increasing levels of serum IgG and detection
or 1 to 10/100 monocytes). No Ep was seen in the of increased RBC-bound IgG by sensitive tests. Two
blood smear of 25 normal controls or nine splenec- patients had serum IgG levels above 1800 mg/dL; the
tomized subjects. The mean hematocrit value of the IgG of one patient was 1950 mg/dL and of the other,
Ep(+) patients was significantly lower than that of the 3000 mg/dL. Both of these patients appeared to have
Ep(–) SCD patients. increased RBC-bound IgG, as indicated by positive sen-
Galili and coworkers,53 using a modified AGT based sitive tests; it is possible that these results were due to
on the high affinity between the Fc portion of RBC- nonspecific uptake of IgG onto the RBCs.
bound IgG and the Fc receptor on the myeloid cell K- Thus, there appeared to be no correlation between
562, also found increased levels of IgG on RBCs from positive direct ELATs, direct Polybrene tests, or reac-
thalassemias. Immunoglobulin was found on the RBCs tive concentrated eluates and serum IgG levels among
of 73 out of 80 patients (90%) with thalassemia. The patients with normal (even up to 1800 mg/dL) IgG
immunoglobulin on the thalassemic RBCs belonged to levels.56 We think it reasonable to continue assuming
the IgG class and was said to be autoreactive. Elution that we are detecting IgG autoantibody rather than
328 Immune Hemolytic Anemias

nonspecifically bound IgG on the RBCs of these RBCs from six normal donors tested at the same time
patients. When rare patients with DAT-negative the patients’ RBCs were tested. The patients with
AIHA and hypergammaglobulinemia (such as the hemolytic anemia obviously have increased amounts
latter two patients just mentioned) are encountered, it of RBC-bound IgG compared with normal donors, but
is always possible that positive sensitive test results it is hard to be convinced that this represents a purely
could reflect nonspecific uptake rather than autoanti- quantitative difference.
body. Thus, to improve the accuracy of the interpreta- When using ELAT, positive controls of IgG- (anti-D)
tion, we would recommend that hematologists relate sensitized RBCs are always tested; Garratty15 noticed
the serum IgG level to the results of sensitive tests for that the ODs on these controls, with obvious positive
RBC-bound IgG. In patients with hypergammaglo- IATs, were often less than the ODs on some DAT-neg-
bulinemia, positive results of tests that are more ative patients. The 1⁄2+, 1+, and 2+ results in Figure 9-3
sensitive than the antiglobulin test would have to be represent the IAT results of RBCs that were sensitized
suspected to be unreliable. with anti-D in vitro and yielded similar ODs to the
DAT-negative patients, which are represented by the
triangle next to the plus (+) signs. Thus, it is a mystery
Does Quantity of RBC-Bound IgG Always why the RBC-bound IgG on the RBCs of the patients
Explain the Phenomenon? Do Qualitative with hemolytic anemia was not detected by the AGT,
Factors Play a Role? when anti-D sensitized cells with similar ELAT results
yielded obvious positive IATs.
Some findings suggest that the explanation for DAT- The results suggest something other than a simple
negative AIHA is not simply quantitative and that it quantitative reason for the detection of RBC-bound
could represent a qualitative defect of the sensitivity of IgG in the presence of a negative DAT. The answer
the routine DAT. Gilliland and associates8,9 and the cannot be differences in the AGS, as Petz and
current authors6 found that some patients classified as Garratty6 used the same anti-IgG for the CFAC tests
having DAT-negative AIHA had far more than 200 IgG that was used for the DATs. It is possible that the RBC-
molecules per RBC. For example, five of the 27 patients bound IgG could be oriented in such a way on the
in our series had more than 400 IgG molecules per RBC RBC membrane that the anti-IgG could not bridge
when measured by the CFAC test (see Table 9-3). IgG between adjacent RBCs to cause agglutination, and
sensitization to this degree should be easily detectable that this IgG would be detected by assays that are not
by the AGT; indeed, more than 400 molecules would dependent on agglutination (e.g., the CFAC test or
usually yield >3+ DAT reactions. ELAT). This might occur if the IgG was not bound to
Garratty15 reported data supporting suspicions that the RBC by its Fab portion (i.e., if it was not an anti-
the explanation might not be totally quantitative. body directed against RBC antigen).
Figure 9-3 shows examples of RBCs tested by ELAT. Engelfriet and colleagues57 suggested that DAT-neg-
The triangles represent the optical density results ative AIHA could be due to small amounts of RBC-
obtained on the RBCs of 12 patients suspected of bound IgG3 autoantibody. In vitro experiments
having DAT-negative AIHA; the circles represent the suggested that about 500–1000 IgG1 molecules per RBC
mean optical density readings (ODs) obtained on the were needed for in vitro interactions with peripheral

DIRECT ELAT

0.5
Patient (mean) 1+
FIGURE 9-3. Comparison of enzyme-linked antiglobu-
Normal RBC (mean)
lin test results on RBCs from patients with “DAT-
0.4 1/2+, 1+, 2+: Equivalent DAT 1 1/2+ 2+ negative” AIHA (▲), normal donors (•) (mean OD of
Optical density

on in vitro lgG (Rh) sensitized 6 donors), and D + RBCs sensitized in vitro with dilu-
RBCs on that day 2+
tions of anti-D and giving the same OD readings as
0.3 the RBCs from the “DAT-negative” patients tested
on the same day. The results of the indirect
1+ 1/2+ antiglobulin tests of the anti-D sensitized RBCs are
graded from 0 to 2 +. (From Garratty G: Factors
0.2 0 affecting the pathogenicity of red cell auto- and
1+
1/2+ alloantibodies. In Nance SJ (ed): Immune
1/2+
1/2+ 1+ Destruction of Red Blood Cells. Arlington, Va:
0.1 American Association of Blood Banks, 1989:
109–169.)

By Ni Ba He Br Kn Wo Ok Ra Pr Go Fr
“DAT negative” AIHA patients
Unusual Aspects of Acquired Immune Hemolytic Anemias 329

blood monocytes, but only 100 to 150 IgG3 molecules DAT-positive RBC samples were subjected to the
per RBC were necessary for similar interactions.57,58 It same conditions, DAT titration scores appeared
should be emphasized that these experiments used similar to those of the RBCs that were tested immedi-
peripheral blood monocytes and that smaller numbers ately following washing; none of the positive DATs
of IgG molecules could yield similar interactions with became negative.
macrophages, which are more active than monocytes. Sokol and coworkers61 described two patients with
Engelfriet and colleagues57 used dilutions of IgG1 suspected AIHA who had negative routine DATs. Their
and IgG3 to sensitize RBCs in vitro; the sensitized DATs were positive when the patients’ RBCs were
RBCs were tested by AGT and a monocyte antibody- washed with saline at 4°C. In one case, the DAT was
dependent cellular cytotoxicity (ADCC) assay. They also positive by a column agglutination technique (ID
found that cytotoxic damage of IgG1-sensitized RBCs gel from DiaMed-AG, Cressier sur Morat, Switzerland).
occurred only when the AGT was positive, but that Eluates from the patients’ RBCs were reactive if eluates
RBCs sensitized with two dilutions of IgG3 anti-D were prepared from RBCs washed at 4°C.
yielding negative AGTs were cytotoxically damaged. Lai and colleagues62 described a patient with
Zupanska and coworkers58 also showed that RBCs hemolytic anemia who had a negative routine DAT
sensitized in vitro with IgG3 anti-D (but not IgG1 anti- but whose DAT was 4+ when the RBCs were washed
D) sometimes adhered to monocytes in an in vitro with saline at 4°C. The DAT was also positive when
monocyte monolayer assay (MMA) when the DAT on tested by column agglutination techniques (CAT).
those RBCs was negative. Thus, it is possible that the Johnson and Bandouveres63 described a 6-month-
difference between normal RBC destruction by natu- old infant with a severe hemolytic anemia (hemoglo-
rally occurring IgG autoantibodies and the abnormal bin of 4 g/dL). The routine DAT was negative. The
destruction associated with DAT-negative AIHA DAT was 3+ after the patient’s RBCs were washed
could be qualitative as well as (or instead of) quanti- with 4°C LISS. An eluate from the patient’s RBCs
tative. It would be of great interest to know whether reacted at 3+, showing anti-Pr specificity. The
the IgG antibody to the RBC senescence antigen (see patient’s serum contained an IgG anti-Pr detectable
Chapter 4) is always IgG1, which would suggest that only by IAT using 4°C LISS for the washing phase.
subclass difference could be more important than the We believe that low-affinity antibodies can some-
subtle quantitative differences. times cause technical problems in obtaining an accu-
rate DAT result; they might possibly cause a
false-negative IAT in addition to a false-negative DAT.
DAT-NEGATIVE AIHA ASSOCIATED WITH Low-affinity autoantibodies (and possibly alloanti-
LOW-AFFINITY IgG AUTOANTIBODIES bodies) may be dissociated from the RBCs by the
process of washing for the DAT (and probably for the
When the routine DAT is performed, the patient’s IAT). This loss of antibody occurs more frequently
RBCs are washed three to four times in large volumes when the RBCs are washed at 37°C (note that 6 of 17
of saline at room temperature (RT). Sometimes, patients referred as DAT negative AIHA were DAT
because cold autoagglutinins are noted, the RBCs are positive when the DAT was performed using the
washed with 37°C saline. We have encountered rare routine method using RT saline). The referring hospi-
patients in whom a DAT that is negative or weakly tals usually had used a “prewarm” procedure.41,59,60
positive becomes strongly positive when the routine It is of interest to add that some of the cases that we
procedure is processed under different conditions. have encountered that were associated with low-
Garratty and associates41,59,60 studied 22 patients affinity autoantibodies have had quite severe
with AIHA who were associated with low-affinity hemolytic anemias. This fact does not seem to agree
autoantibodies; 17 of these patients were referred with early published work that RBCs sensitized with
because of suspected DAT-negative AIHA. Eleven of high-affinity antibody are destroyed much more
the 22 (including 6 of 17 referred as DAT-negative efficiently than those sensitized with low-affinity anti-
AIHA) patients had a positive DAT if the RBCs were bodies (see also Chapter 4).15,41,64
washed with saline at RT, but the DATs were negative Schwartz and Costea64 performed the only pub-
in 8 of 11 patients when the RBCs were washed at lished experiments relating alloantibody affinity to in
37°C. RBC-bound IgG was detected on all 22 patients vivo RBC destruction. They studied three examples of
when the RBCs were washed with ice-cold (0–4°C) anti-D with different affinities for D+ RBCs. They
saline and/or low ionic-strength saline (LISS). When expressed the antibody affinity in terms of the amount
the RBCs were washed with cold saline or LISS, they of time it took for in vitro spontaneous elution of 50%
were always tested with 10% albumin (as a negative of the RBC-bound anti-D. The elution times (ET) were
control), in parallel with the antiglobulin sera, to 160 minutes, 100 minutes, and 40 minutes, respec-
control against cold autoagglutinins causing the posi- tively, for the three anti-D. RBC survival studies were
tive test result. When RBCs from some of these carried out on 51Cr-labeled RBCs sensitized with these
patients were washed and allowed to stand at RT for three anti-D. Figure 9-4 shows the results. There was
20–160 minutes before performing the DAT, all DATs an inverse relationship between RBC survival and ET;
were much weaker or negative. When 22 random that is, the RBCs coated with anti-D of the highest
330 Immune Hemolytic Anemias

100 showed that low-affinity autoantibodies in a mouse


model had high pathogenic potential.65
Although low-affinity antibodies appear to be
uncommon, the foregoing results emphasize that
50 Anti D (1) ET = 40⬚ when we wash RBCs, we are creating conditions very
different from those found in vivo. The antiglobulin
test must be performed immediately following
washing of the RBCs. If RBCs are washed at 37°C, one
Anti D (2) ET = 100⬚ should remember that some antiglobulin tests could be
weaker or even negative when low-affinity antibodies
are present; on rare occasions, even RBCs washed at
% CPM

RT might yield false weak positive or negative


antiglobulin tests.
10

DAT-NEGATIVE AIHA ASSOCIATED


5
Anti D (3) ET = 160⬚
WITH RBC-BOUND IgA AND IgM

Routine polyspecific AGSs are not standardized to


react with IgA- or IgM-sensitized RBCs; they are
4 8 12 16 20 24 required only to detect IgG and C3 (C3d). Most of the
Hours polyclonal polyspecific AGS used routinely for DATs
FIGURE 9-4. The effect of antibody affinity on RBC survival: The ordi- do contain anti-IgA, but they sometimes react only fol-
nate and abscissa indicate the in vivo RBC survival of D + 51Cr-labeled lowing the 5- to 10-minute incubation period recom-
RBCs sensitized with three anti-D, of different affinity, in terms of 51Cr mended by the manufacturers to enhance anti-C3
counts (% CPM) and collection time of samples tested, respectively. ET reactivity.66 Anti-IgA and anti-IgM that are standard-
refers to “elution time,” which is the time required for 50% of the sen-
sitizing antibody to dissociate from the RBCs In vitro. Thus, short
ized for the DAT are not readily available. One can
elution time indicates a low avidity antibody [e.g., anti-D (1)], and a long standardize immunological reagents, but as agglutina-
elution time indicates an antibody with higher affinity [e.g., anti-D (3)]. tion is far more sensitive than many immunological
(From Schwartz RS, Costea N: Autoimmune hemolytic anemia: Clinical procedures, great care must be taken to avoid
correlations and biological implications. Semin Hematol 1966;3:2–26.) specificity problems with heterophile antibodies and
anti-light chain cross-reactivity (see Chapter 6). Anti-
IgM does not work well with the DAT, even when a
reagent that is potent by other criteria is used.6 We
affinity (longest ET) had a shorter survival than the
(and others) have been quite successful detecting RBC-
RBCs coated with anti-D of lower affinity (shorter ET).
bound IgA and IgM using flow cytometry.41,42,67-70
Although equilibrium constants were not calculated,
On rare occasions, WAIHA can be associated with
one would expect that the ET, as measured in these
IgA and IgM autoantibodies without the presence of
experiments, should correlate with the equilibrium
IgG. For the reasons just discussed, they can some-
constants of the antibodies. These experiments were
times present as DAT-negative AIHA.
carried out in 1966, and it is surprising that there are
no other good data in humans relating antibody
affinity to the degree of RBC destruction; this relation- WAIHA Associated with IgA
ship would appear to be an important factor. Autoantibodies
Garratty15 suggested that one explanation might be
that the low-affinity antibodies that were detected The clinical and hematological features of patients with
might not have been the major cause of the severe AIHA associated with IgA autoantibodies are very
hemolysis in the patients discussed previously. High- similar to those of patients whose AIHA is associated
affinity antibodies might have been present and, with warm IgG autoantibodies.66,70-83 Patients with
being of higher affinity, would have reacted with the both types of AIHA also respond to the same therapy
patient’s RBCs first, leaving predominantly lower- (i.e., steroids and/or splenectomy). Macrophages were
affinity antibodies in the plasma. If most of the RBCs originally said not to have (or to have very inefficient)
sensitized with the high-affinity antibody were receptors for IgA, but increasing evidence suggested
removed by the mononuclear phagocyte system, the that IgA sometimes participates in phagocytosis and
only RBCs left circulating might be those sensitized ADCC.84-87 A human Fc receptor (FcαR) for IgA has
with low-affinity autoantibodies. This is a good been cloned.84 The monoclonal antibody used to isolate
example of where the results of our in vitro tests the clone is capable of inhibiting any binding of IgA
might not correlate with the clinical course observed and phagocytosis of IgA-coated targets.88
in actual patients. In contrast to the early experiments Clark and associates75 studied the mechanism of
using human antibodies, a more recent publication hemolysis in a patient whose WAIHA was associated
Unusual Aspects of Acquired Immune Hemolytic Anemias 331

with an IgA autoantibody. The patient pursued a clin- with IgM autoantibodies had a negative DAT when
ical course (spherocytosis, splenic sequestration of routine AGS (anti-IgG and anti-C3) were used; IgM
red cells, response to corticosteroids, and splenec- was detectable on the RBCs of two of these four
tomy) very similar to that of individuals with patients using flow cytometry. Sokol and coworkers34
WAIHA associated with IgG autoantibodies, which studied 219 patients suspected of having AIHA. DAT
suggests that the mechanism of hemolysis might be and ELAT results were the same in 61 patients; in 43
similar to that occurring in WAIHA associated with cases, the ELAT detected additional immunoglobulin
IgG autoantibodies. An eluate from the patient’s classes; in 33 cases, the DAT showed only RBC-bound
RBCs was shown to react by ADCC toward sensitized C3 (C3d), but ELAT showed RBC-bound IgM in 16
RBCs that appeared to be IgA dependent. The ADCC- (50%) of these. In five of 16 cases, the DAT was negative
promoting effect of the eluate was blocked by a solid- and the ELAT detected RBC-bound IgM. Kay and asso-
phase anti-IgA immunosorbent and by fluid-phase ciates89 and Szymanski and colleagues92 detected low
normal IgA, and the effect was dependent on the molecular weight (monomeric) IgM on patients’ RBCs.
dose of IgA eluate used to sensitize RBCs. The It is still controversial whether IgM non–complement-
patient’s eluate promoted phagocytosis of sensitized activating antibodies can cause immune RBC des-
RBCs by normal mononuclear phagocytes. Both truction, and if they do, how this is possible, as
ADCC and phagocytosis promoted by the patient’s macrophages are said to lack IgM receptors.
eluate were complement independent. The eluate Schreiber and Frank93,94 injected a strain of guinea
failed to produce complement-mediated hemolysis, pig, genetically deficient in C4, with 51Cr-labeled
and complement components could not be detected on IgM sensitized RBCs (it should be noted that these
sensitized RBCs either in vitro or in vivo. Taken RBCs were sensitized with relatively small numbers
together, these observations suggested a possible of IgM molecules) and showed that the RBCs sur-
mechanism for what appeared, clinically, in this patient vived normally. Later, similar experiments in
to be a WAIHA mediated by IgA—a complement- humans with complement deficiencies yielded
independent interaction of IgA-coated RBCs with the similar results.95 IgM anti-M, anti-D, and anti-c have
patient’s mononuclear phagocytes. been reported as a cause of increased RBC destruc-
Salama and colleagues78 studied a 6-year-old tion; it was suggested that RBC agglutinates might
patient with AIHA who had a strongly positive DAT become trapped in small blood vessels and even-
due predominately to IgA, although small amounts of tually suffer metabolic damage.96,97 Mollison98,99
RBC-bound IgG were also detected; no RBC-bound reviewed his earlier data and reported that he could
C3 was detected. This patient had severe hemolysis not be confident that the IgM anti-M studied in
associated with an upper respiratory tract infection. 195896 did not activate complement. He also could
Evidence was presented that C3-independent intra- not exclude the possibility that some IgG anti-D had
vascular immune hemolysis had occurred through been present, in addition to the IgM anti-D, in the
“reactive hemolysis” (see the section on bystander serum reported in 1971.97 He had no good explana-
immune cytolysis later in this chapter). Reactive tion for the increased destruction of the RBCs that
hemolysis involved C3-independent binding of C5b-9 were seemingly sensitized only by IgM anti-c.97 In this
complexes in addition to IgA (and IgG), but no C3. case, 51Cr-labeled c+ RBCs were almost totally cleared
Salama and colleagues78 pointed out that several in the liver, which suggested complement-mediated
reports of AIHA associated with IgA autoantibodies destruction rather than IgG-mediated destruction, but
appeared to have signs of intravascular lysis and that no in vitro complement sensitization was observed.99
perhaps reactive lysis might play a role in the Thus, there would seem to be very little experimen-
hemolytic process. tal evidence that RBC-bound IgM alone can cause
increased RBC destruction. Nevertheless, there are
WAIHA Associated with IgM reports of cases of AIHA that appear to be associated
Autoantibodies with non–complement-binding IgM antibodies.
Salama and Mueller-Eckhardt100 reported on 12 chil-
IgM warm autoantibodies might agglutinate untreated dren (eight of whom were infants) with relatively
and/or enzyme-treated RBCs, hemolyze untreated severe AIHA associated with non–complement-
and/or enzyme-treated RBCs, and/or sensitize RBCs binding IgM autoantibodies. The routine DATs were
so that IgM and/or complement is detectable by the negative in 11 cases and positive due to RBC-bound
DAT.67-69,89-92 The routine DAT is sometimes negative C3d in one case. All patients were shown to have
in patients with warm IgM warm autoantibodies, RBC-bound IgM when a radioimmune assay was
however. used; two cases also had IgA and/or IgG detected by
Some authors have detected IgM on the RBCs of this assay. An unusual finding with these patients was
patients who have AIHA but a negative routine DAT. that no antibodies were detected in the patients’ sera
67-69,89-92 This IgM was sometimes detectable by the either by conventional serology or the radioimmune
DAT using anti-IgM, and sometimes detectable only by assay. Eluates from RBCs of all 11 patients were non-
more sensitive procedures. Garratty and colleagues69 reactive by conventional serology, but two reacted
found that four of 41 (10%) cases of WAIHA associated with anti-IgM by the radioimmune assay.
332 Immune Hemolytic Anemias

(For a further discussion of AIHA associated with routine RT wash, we wash the patient’s RBCs with
warm IgM autoantibodies, see Chapter 5.) cold LISS or saline. Careful controls are necessary to
ensure that false-positive DATs are not caused by cold
SEROLOGICAL AIDS IN DIAGNOSING autoagglutinins (see Chapter 6). If cold agglutinins are
a problem with a 0–4°C wash, then washing with RT
DAT-NEGATIVE AIHA LISS could be a compromise.

The sensitive assays discussed previously for detecting


RBC-bound IgG below the threshold of the DAT are
Using Column Agglutination Tests
sometimes called by hematologists “Super Coombs’ (CAT) for DAT
tests.” These assays are tedious to standardize and CATs (e.g., gel test) sometimes yield a positive DAT
perform; they often take most of a working day to on RBCs that are DAT negative by routine tube
obtain a result. Their predictive value is not good. methods.62,101-103 This could be because no washing of
Garratty and colleagues40,41 reported that less than one RBCs is involved; thus, low-affinity autoantibodies
third of DAT-negative patients suspected of having that would be lost during the wash phase used in the
WAIHA have increased amounts of RBC-bound IgG tube method can be detected.
detected by such assays (Table 9-5). These assays are
not available in many laboratories in the United States;
even in our own laboratory, we rarely employ them. Using Special Antiglobulin Sera
Our routine approach now is to employ serological
We test patients’ RBCs with anti-IgA and anti-IgM
tests, such as the ones listed later in this chapter;
that have been standardized for use with RBCs (see
methods are given in more detail in Chapter 6.
Chapter 6). We also use an anti-C3 that is known to be
rich in anti-C3d.
Direct Antiglobulin Tests
We sometimes find that a DAT reported as negative by
a referring hospital is weakly positive, with routine
Direct Polybrene Test
reagents, when tested by experienced technologists in We have found that one of the most useful serological
our reference or research laboratories. In one study, tests for detecting RBC-bound IgG that is undetectable
Garratty and coworkers40 found that 10% of DATs by routine DAT is the direct Polybrene test (see Chapter
reported as negative by referring hospitals were posi- 6 for details of performing the test).28,40,41,104 Normal
tive when tested in their research laboratory. The RBCs aggregate when added to Polybrene in a low-
false-negative results obtained by the hospital some- ionic strength milieu. This aggregation can be dis-
times occurred because the hospital unnecessarily persed by adding sodium citrate and mixing gently. If
washed the patient’s RBCs at 37°C; loss of RBC- RBCs are coated with IgG added to Polybrene and cen-
bound, low-affinity IgG warm autoantibodies is trifuged, they agglutinate, and this agglutination is not
greater when RBCs are washed at 37°C.41,59,60 dispersed by sodium citrate. If agglutination is not
observed at this stage, RBC-bound IgG can sometimes
Washing Patient’s RBCs in Room be detected by washing the RBCs and adding anti-IgG.
Temperature LISS or Cold (0–4°C) Such a test can detect RBC-bound IgG that is not
Saline or LISS detectable by routine DAT. Garratty and colleagues28,40
found the direct Polybrene test to be almost as sensitive
To retain low-affinity RBC-bound autoantibodies as flow cytometry and more sensitive than ELAT
during the washing phase, in addition to using the (which is equivalent to the CFAC test) for detecting
RBC-bound IgG (see Table 9-5). The method we use is
described in detail in Chapter 6. Owen and Hows105
TABLE 9-5. COMPARISON OF TESTS USED FOR and Rubino and associates,106 using a slight variation of
DETECTING RBC-BOUND IgG WHEN DAT IS NEGATIVE the procedure given in Chapter 6, also found the direct
Polybrene test useful.
No. of No./% If Polybrene is not available, a polyethylene glycol
Test Patients Positive (PEG) DAT can be used, but Garratty and colleagues40
Flow cytometry 70 15/21
and Rubino and coworkers106 did not find this as sen-
Direct Polybrene 205 38/19 sitive as the Polybrene test.
Direct ELAT 201 32/16
Direct PEG 160 16/10
Concentrated eluate 162 15/9 Detection of Autoantibody in Eluates
DAT using cold saline washes 209 7/3
MMA 105 4/4 Gilliland and associates8 found that they could
demonstrate that IgG autoantibody was present on the
From Garratty G: Autoimmune hemolytic anemia. In Garratty G (ed):
Immunolobiology of Transfusion Medicine. New York: Dekker, 1994: RBCs of six of six patients with DAT-negative AIHA if
493–521. they tested concentrated eluates prepared from the
Unusual Aspects of Acquired Immune Hemolytic Anemias 333

patients’ RBCs. Eluates (three cycles) were prepared patients without increased RBC destruction. Galili
from 50–200 mL of a patient’s RBCs and then concen- and associates112 used an assay measuring adherence
trated to 1 mL. The eluates reacted strongly by the of patients’ RBCs, following incubation in antiglobu-
routine IAT. It is not often possible or practical to use lin serum, to a K-562 erythro-myeloid cell line. Six of
this approach, but we also have found that eluates pre- six DAT-negative patients suspected of having AIHA
pared by a routine method from 3–5 mL of a patient’s gave positive results with this assay.
RBCs will sometimes react (see Table 9-5).6,28,40-41 We Nance and Garratty113 used an MMA that mea-
found that a two- to fivefold concentration yielded sured both RBC adherence and phagocytosis. RBCs
more positive reactions (we used Minicon filters from 174 normal donors with negative DATs had a
[Amicon Corporation, Lexington, MA] to concentrate mean total reactivity (MTR) of adherence and
the eluates). phagocytosis of 0.3%. RBCs from 45 normal donors
with positive DATs (mean of 2+) had an MTR of
0.6%. They had definitive clinical and laboratory
Detection of IgG Autoantibodies data on 44 of 70 DAT-positive patients tested.
in Patients’ Sera Twenty-five of the 44 had no evidence of hemolysis
at the time of testing and had an MTR of 3.8%,
Sometimes IgG autoantibodies are readily detectable
whereas the 19 with AIHA had an MTR of 11.4%.
in the patient’s sera when the DAT is negative.6,107-109
The MTR of RBCs from 19 other patients with DAT-
These autoantibodies are usually potentiator
negative AIHA were within two standard deviations
dependent, only being detectable with the use of
of the normal range. The MMA results correlated
enzyme-treated RBCs, in the presence of PEG, or by
perfectly with the presence of AIHA, except in one
using CATs. The results might indicate the presence
patient with AIHA and a 2+ DAT, whose RBCs
of low-affinity antibodies that are lost from the
yielded 0% reactivity in the MMA. The MTR of DAT-
patient’s RBCs during the washing phase of the IAT,
positive patients with AIHA was three times greater
or transient depression of antigens on patients’
than that of DAT-positive patients without AIHA,
RBCs108,109 (see also Chapter 7).
19 times greater than that of DAT-positive normal
Table 9-6 summarizes the laboratory tests we per-
donors, and 42 times greater than that of DAT-
form to evaluate a patient referred to our laboratory
negative normal donors. Later results by the same
for DAT-negative AIHA.
laboratory found that RBCs from only four of
105 patients with DAT-negative AIHA reacted by the
CORRELATIONS WITH IN VITRO MMA (see Table 9-5).40
Gallagher and associates114 also found a good cor-
FUNCTIONAL CELLULAR ASSAYS relation with their assay and AIHA. RBCs from all
18 DAT-positive patients with AIHA adhered or were
The first report we know of applying cellular assays phagocytosed by monocytes. RBCs from all 10 DAT-
to investigate DAT-negative AIHA is that of Parker positive patients without AIHA were not significantly
and coworkers110 in 1972. These investigators used an phagocytosed, but four of the 10 showed increased
erythrophagocytosis assay, in which a patient’s adherence to monocytes. This led Gallagher and
washed RBCs were added to mouse macrophages. associates114 to use only phagocytosis as an index of
The assay was strongly positive in a patient who was clinical significance in future studies. The same reasear-
DAT negative but appeared to have AIHA. van der chers also studied 11 patients with DAT-negative AIHA.
Meulen and colleagues111 also applied an RBC adher- RBCs from seven of the 11 patients yielded significantly
ence assay to study RBCs from patients with AIHA. elevated results; however, three of these results were
RBCs from 42 patients with positive DATs were significantly elevated only if autologous monocytes
added to monocytes in vitro, and RBC adherence were used. Of the seven patients showing significant
occurred with RBCs from all 22 patients who had reactions, six had a weakly positive DAT using C3d; it is
AIHA; no adherence was noted with RBCs from 20 unclear whether this influenced the reactions.

TABLE 9-6. FURTHER TESTING OF RBCs FROM PATIENTS WHO HAVE A NEGATIVE DAT AND SUSPECTED AIHA

1. Repeat DAT. If referral laboratory washed RBCs at 37°C, it is important to use RT washing when the DAT is repeated.
2. DAT using anti-IgA and IgM in addition to anti-IgG and anti-C3; it is important that the anti-C3 contain potent anti-C3d activity.
3. If DAT is still negative, repeat DAT using RBCs washed with 4°C saline or, preferably, 4°C LISS (controls to exclude false-positive results due to
cold auto-agglutinins are essential—see Chapter 6).
4. Direct Polybrene test (see Chapter 6 for details).
5. Additional serological tests that might be useful include a DAT using CAT (e.g., gel columns); IAT using patient’s serum and/or an eluate from
the patient’s RBCs, tested against enzyme-treated RBCs or in the presence of PEG.
6. If serological tests are nonproductive, a DAT using flow cytometry (or one of the other sensitive assays discussed earlier) can be used.
334 Immune Hemolytic Anemias

Brojer and coworkers115 also found a good correla- twice for symptomatic anemia. The response to
tion with their monocyte assays and in vivo hemolysis transfusions was good.
in DAT-positive patients. The same group found that To assess a possible role for NK cells in the hemol-
the number of RBC-bound IgG molecules generally ysis, a cytotoxicity assay was designed.119 NK cells
correlated with the presence of phagocytosis and the were obtained by density gradient and purified.
degree of anemia, but there were exceptions.116 Cytotoxicity was measured by a 51Cr-specific release
Phagocytosis was always observed when there were assay using autologous RBCs and ABO-identical
more than 2000 IgG molecules per RBC. In patients allogeneic RBCs as targets. The resulting hemolysis
having less than 700 IgG molecules per RBC, the inter- was expressed as a percentage of the mean release,
action with monocytes was observed only if IgG3 was corrected for spontaneous release. Autologous RBCs
present on the RBCs; the minimum number of IgG3 (but not allogeneic RBCs) were lysed in vitro by the
molecules per RBC initiating phagocytosis was patient’s NK cells, either in the presence or absence
150–640, in contrast to RBCs with IgG1 sensitization of serum, which indicated that a direct lytic mecha-
where 1250 to 4020 molecules were needed. They nism was effected by the proliferating and activated
found no differences in reactivity when comparing NK cells. The patient was started on treatment with
autologous monocytes with homologous monocytes cyclophosphamide. After 1 month, the anemia
from patients with AIHA.116 improved, and 2 months later, NK blood cell counts
Using a mononuclear phagocyte assay, Herron and and spleen size normalized. Thereafter, the patient’s
colleagues117 found a good correlation with AIHA but NK cells failed to show any lytic activity against
failed to detect RBC-bound IgG on three DAT- autologous RBCs in the cytotoxicity assay. It was
negative patients suspected of having AIHA. suggested that NK cell activation and cytotoxicity
In summary, although on occasion we have could have contributed to the hemolytic anemia in
obtained a positive result (sometimes with only auto- this patient with large granular lymphocytic
logous monocytes), in many cases the MMA was non- leukemia.119
informative. We no longer use the MMA routinely in
our workup of DAT-negative AIHA.
THERAPY AND COURSE
Could DAT-Negative AIHA Be Due to an
Antibody-Independent, Cell-Mediated, Patients with acquired hemolytic anemia who have a
negative DAT without evidence for a nonimmune
Cytotoxicity Mechanism? cause of the hemolysis, but who do have abnormal
In 1981, Garratty118 suggested the possibility that numbers of IgG molecules on their RBCs, should be
natural killer (NK) cells might recognize foreign treated similarly to patients who have more charac-
determinants on RBCs and cause alloimmune or teristic findings of WAIHA (see Chapter 11). The
autoimmune hemolytic anemia by antibody-inde- variable responses to therapy indicated in Table 9-3
pendent cell-mediated cellular cytotoxicity. As six of and described previously are similar to these
22 patients with hemolytic transfusion reactions and observed in patients who present with a positive
no demonstrable antibodies had chronic lymphatic DAT and typical serologic findings of WAIHA.
leukemia, this author tried to demonstrate NK cell Gilliland9 has also demonstrated that, in many
activity by one chronic lymphatic leukemia patient’s patients, a reduction of cell-bound IgG antibody can
mononuclear cells against putative donor RBCs; the be documented after remission induced by corticos-
experiment was unsuccessful. teroids or splenectomy. Other patients, however,
Gilsanz and colleagues119 reported success in develop a well-compensated hemolytic anemia with
demonstrating NK cell activity against autologous little change in the abnormal quantity of IgG per
RBCs in a patient with DAT-negative AIHA. A 65- RBC. Gilliland further indicated that some patients
year-old patient with a 5-year history of asympto- who originally presented with AIHA with a positive
matic large granular lymphocytic leukemia DAT, once given corticosteroids therapy, have shown
developed hemolytic anemia. She had a hemoglobin a reduction of RBC-bound IgG antibody to levels
of 97 g/L and an increased reticulocyte count (6%); that are still elevated, but below detection by routine
the bilirubin was 1.4 mg/dL and lactate dehydroge- antiglobulin testing.9 The quantity of IgG antibody in
nase (LDH) was 350 IU/L; serum haptoglobins were some of these patients remained at these low levels
absent. The peripheral blood smear showed marked for several years, whereas in others, the quantity of
spherocytosis; splenomegaly was found by ultra- IgG per RBC increased, and hemolytic anemia
sonography. DATs were repeatedly negative. The recurred after therapy with steroids was decreased
large granular lymphocyte count was 6.0 × 109/L; the or stopped. In still other patients, the quantity of IgG
NK phenotype was CD2+, CD3–, CD16+, CD38+, per RBC fell to the range observed on normal RBCs,
CD57+. No coexisting disease or hemolytic trigger- and therapy was discontinued without an increase of
ing factors were found. She did not improve with RBC-bound antibody or exacerbation of hemolytic
prednisone (20 mg/day) and had to be transfused disease.
Unusual Aspects of Acquired Immune Hemolytic Anemias 335

B. Development of RBC Autoantibodies


and AIHA Following Transfusion

Our interest in the role of blood transfusion as a possi- tinins. In 1965, Ovary and Spiegelman121 confirmed
ble cause of AIHA was stimulated by the observation and extended these findings. These authors found
that one of our patients who developed two RBC that IgG warm autoantibodies sometimes were
alloantibodies also developed a positive DAT and formed in addition to the IgM cold autoagglutinins. In
AIHA after receiving blood transfusions.6 The patient the same year, Zmijewski122 found that chimpanzees
was a group O Rh-negative elderly male with gastroin- developed autoagglutinins after immunization with
testinal bleeding of undetermined origin; he received human RBCs or leukocytes. The chimpanzees also
two units of group O Rh-positive RBCs. The DAT and developed strongly positive DATs due to RBC-bound
IATs were negative at this time. Eight weeks later, anti- IgG and complement. The autoantibody(ies) eluted
D was present in his serum, but the DAT remained from the RBCs reacted with human RBCs and with
negative. Two units of group O Rh-negative RBCs were some chimpanzee RBCs. Adsorption of the eluate
transfused, and subsequently, no further transfusions with human RBCs removed all activity; however,
were required. Two months later, anti-D (titer of 1024), adsorption with chimpanzee RBCs removed activity
anti-E (titer of 8), and anti-c (titer of 8) were detected in for chimpanzee RBCs but left activity for human
his serum, as was a weak IgG “nonspecific” antibody RBCs. Zmijewski122 suggested that the phenomenon
that reacted with all red cells of a panel. The DAT was was due to cross-reacting antibodies. Liu and Evans123
positive, and IgG antibody eluted from the RBCs also had described the occurrence of a positive DAT in
showed no evident blood group specificity. Three three of 12 rabbits following intraperitoneal injections
weeks later, the DAT remained positive, the “non- of allogeneic blood. These experiments were per-
specific” autoantibody in the serum had increased in formed to simulate the observation of a transient pos-
titer, and the patient had developed mild signs of itive DAT in a patient with anemia secondary to
hemolytic anemia. The DAT was performed biweekly intraperitoneal bleeding from a ruptured ectopic preg-
for the next 4 months and remained positive without nancy. It was suggested that relatively bland treat-
any diminution in the strength of the reaction. Signs of ment of RBCs could result in modification of surface
hemolysis gradually subsided spontaneously, and per- structure sufficiently to produce antibody formation.
formance of the DAT monthly indicated a gradual There are several publications relating to the induc-
weakening of the reaction during the subsequent 6 tion of RBC autoantibodies in mice immunized with
months. The patient was next evaluated 2 years later, at rat RBCs.124-126 The mice developed autoagglutinins
which time the DAT was negative. (reactive to 37°C) and positive IATs; they also became
Although the development of autoantibodies after DAT-positive. Some mice developed AIHA. It is of
blood transfusion is not recognized frequently, there is great interest that persistence of the positive reactions
a significant number of recorded observations relating (e.g., DAT) required that the mice be immunized
to this phenomenon (Table 9-7). repeatedly with rat RBCs. After immunization was
stopped, the serologic tests returned to normal. When
the mice were immunized again with rat RBCs, there
DATA FROM ANIMAL was a burst of autoantibody production, and then,
EXPERIMENTATION after 4 to 5 weeks, autoantibody production ceased.
Suspensions of lymphoid cells taken from previously
As early as 1918, Rous and Robertson120 showed that immunized mice and transferred to syngeneic normal
rabbits that had received small transfusions of blood mice prevented the induction of autoantibodies when
from other rabbits developed strong cold autoagglu- those mice were injected with rat RBCs. The effect was

TABLE 9-7. EVIDENCE FOR AUTOANTIBODY STIMULATION FOLLOWING TRANSFUSION

Year Reference Recipient Donor Laboratory Findings

1918 120 Rabbit Rabbit “Cold” RBC autoagglutinin


1952 123 Rabbit Rabbit Positive DAT
1965 121 Rabbit Rabbit RBC autoagglutinin; positive IAT
1965 122 Chimpanzee Chimpanzee RBC autoagglutinin; positive IAT; positive DAT (IgG+C3); positive eluate
1973–81 124–126 Mouse Rat RBC autoagglutinin; positive IAT; positive DAT
1978–81 127, 128 Marmoset Marmoset Platelet antibody; platelet-associated IgG
1943–89 129–139 Human Human Positive IAT; positive DAT; reactive eluate
1990–2001 140–143 Human Human Lymphocyte activity/subpopulations
336 Immune Hemolytic Anemias

specific in that it prevented the production of autoan- developed strongly positive DATs, with both autoan-
tibody but not of antibody to rat RBCs. Rats and mice tibodies and alloantibodies present in the serum. The
are known to share common cellular epitopes (e.g., on authors suggested that an explanation for this finding
their RBCs). Thus, it was thought that autoantibody was the lack of control over antibody production that
activity reflected antibody cross-reactivity.124–126 It is is always taking place. They suggested that it might
interesting to note that when RBCs from mice were be assumed that all individuals are subject to RBC
given to rats, a much smaller autoimmune response autoantibody production, but that hemolytic anemia
was seen than that described for mice immunized results only when this “horror autotoxicus” outruns
with rat RBCs.125 normal curbs. They also suggested that immunization
Gengozian and coworkers127,128 described a model to an Rh structure common to all Rh antigens could
of post-transfusion purpura (PTP) in marmosets. yield results suggestive of nonspecificity resembling
When one species of marmoset was immunized with that seen with autoantibodies. Such immunization
platelets from another species of marmoset, thrombo- could arise either de novo at the initial sensitization or
cytopenia developed. This was associated with as a result of initial immunization to a blood factor in
platelet antibody that was detectable in the recipient’s the Rh system, with subsequent widening of the spec-
serum. This antibody reacted with donor platelets but trum of specificity as further stimulation occurred.
did not react (or reacted very weakly) with the recip- Chown and associates133 described two Rh-negative
ient’s platelets. A consistent finding, however, was women who produced Rh antibodies during pregnancy
platelet-associated IgG on the recipient’s platelets. In and later developed positive DATs. Cook134 immunized
those animals that recovered from the thrombocy- 34 Rh-negative male volunteers with Rh-positive blood.
topenia, IgG-coated platelets were shown to be circu- In addition to the development of the expected anti-D in
lating for 30 to greater than 100 days. IgG antibody 68% of the volunteers, 11 men developed an antibody
eluted from these platelets reacted equally well with that reacted with Rh-negative enzyme-treated RBCs;
platelets from other marmosets of the same species as two developed a positive DAT (inhibited by pure IgG)
the recipient or donor. These findings were in contrast that persisted for about four months. No evidence of
to the finding that the serum antibody would react hemolytic anemia was present, although RBC survival
strongly with platelets from marmosets of the same studies were not performed. Beard and coworkers135
species as the donor but reacted only moderately with described an Rh-negative patient who was transfused
platelets of 75% (and weakly with platelets of 25%) of with 400 mL of Rh-positive blood. The foreign cells
marmosets of the same type as the recipient. The were eliminated from the circulation over a period of
weaker reactions, with 25% of the same species, were 5 days by large doses of anti-D immunoglobulin
similar in strength to these observed when autologous assisted by a rapid immune response. Six months later,
platelets taken from marmosets in remission were the patient developed a positive DAT that persisted for
tested. This suggested two populations of antibody, 3 months. Lalezari and colleagues136 described a
one with stronger cross-reactive properties that were 40-year-old black woman who had a partial D pheno-
adsorbed by the recipient’s platelets. type. She had been transfused with several units of Rh-
positive blood, and 10 years later she received three
additional units. She developed a high-titer anti-D, and
DATA FROM HUMANS this anamnestic response was associated with the devel-
opment of an autoantibody. The DAT, which had been
In 1943, Dameshek and Levine129 reported a case in negative prior to transfusion, became positive and
which successive transfusions resulted in extreme, remained so for 6 months. An eluate from the patient’s
almost fatal, hemolysis. They suggested that an irre- RBCs revealed a strong anti-D and a weak “panagglu-
versible autohemolytic process had been established tinin” detected by the Polybrene technique.
by the multiple transfusions and that this was then Worlledge137 has stated that persistent alloimmu-
intensified by a severe hemolytic transfusion reaction nization could lead eventually to autoimmunization,
(HTR). Allen, in discussing a paper presented at the and that one sees patients who have had repeated
Eighth congress of the International Society of Blood transfusions and developed multiple alloantibodies,
Transfusion, stated that he had seen acute acquired in whom positive antiglobulin tests appear even
hemolytic anemia complicating HTRs.130 Polesky and though compatible RBCs have been given. The anti-
Bove131 described a fatal HTR in a woman with body eluted from the RBCs in such cases often seems
marrow hypoplasia associated with acute leukemia. to react with all RBCs except Rhnull cells.
Red cell survival studies, which were in progress at Chaplin and Zarkowsky138 described four patients
the time, made it possible to demonstrate that a severe with SCD who developed AIHA; a fifth patient devel-
autohemolytic crisis was part of the reaction. The oped a positive DAT without AIHA. In the patients
authors postulated the presence of autoantibodies but with AIHA, anemia was severe, with reticulocyte
could not demonstrate their presence. counts of 60%–88%. All five patients had made alloan-
Fudenberg and colleagues132 described two patients tibodies as a result of transfusions before they made
who produced alloantibodies as a result of transfusion autoantibodies. All patients responded to steroid
and then deliberate immunization. Both patients therapy. After discharge, the DATs became negative.
Unusual Aspects of Acquired Immune Hemolytic Anemias 337

Sosler and associates139 described two multiply- bodies in humans but did not involve transfusion of
transfused alloimmunized patients with SCD who donor RBCs. These investigators transfused volun-
developed severe AIHA. One patient had made seven, teers with potent incompatible Rh (anti-CD) antibod-
and the other five, alloantibodies before making ies. Approximately 240 mL of antibody-containing
autoantibodies. Both patients responded to steroids. plasmas were transfused into three recipients of
Argiolu and coworkers140 reported four patients with cDE/cDE, CDe/cDe, and cde/cde phenotypes. Acute
thalassemia major in whom they diagnosed AIHA on hemolytic anemia ensued in the D-positive recipients.
the basis of an increase in transfusion requirement (see DATs on the RBCs were positive immediately, remain-
Chapter 2) associated with the presence of RBC autoan- ing so for 168 and 202 days, respectively, for the
tibodies. All of the patients were treated with intra- cDE/cDE and CDe/CDe recipients. Eluates from the
venous IgG (IVIG) therapy, and three responded as RBCs of the cDE/cDE recipient revealed activity for
indicated by normalization of the blood consumption. his pretransfusion RBCs and cDe/cde RBCs to the end
Chan and associates141 reported two patients who of the study. A surprising finding was the presence of
had been transfused and who later developed auto- a specific anti-E in the RBC eluate. This antibody
antibodies. The relationship between the transfusions appeared on the 70th day after transfusion and per-
and the development of autoantibodies is clouded, sisted at a high level through the 229-day observation
however, by the fact that the first patient had a 9-year period. The authors concluded that the anti-E could
history of chronic lymphocytic leukemia, and that the represent only new autoantibody formation by the
second patient was not evaluated until 4 years after a recipient himself. They hypothesized that the autoan-
prior hospitalization. In both instances, transfusions of tibodies were produced in response to alterations
RBCs that were weakly incompatible because of the brought about at the Rh sites on the RBC membrane
autoantibody were followed by acute hemolysis, which by the antigen-antibody reactions between the D-
was fatal in the second patient. Detailed serologic tests, antigen receptors on the recipient’s RBCs and the
including adsorption, revealed no alloantibodies in transfused anti-CD alloantibodies, involving the E-
either patient’s serum. antigen receptors in the autoimmunization process by
Zumberg and coworkers142 described four patients reason of their proximity.
who developed autoantibodies in temporal association Wodzinski and associates146 described an extraordi-
with alloantibody formation. In two patients, no hemol- nary case of PCH following an ABO-incompatible
ysis developed. One patient, a 25-year-old woman with transfusion. The authors suggested that the ABO HTR
SCD, developed a typical sickle cell HTR syndrome (see triggered or exacerbated an autoimmune response
Chapter 14) characterized by a declining hematocrit and manifesting as PCH.
signs of increased hemolysis following transfusion of
four units of RBCs. The hemolytic reaction was attrib- RETROSPECTIVE REVIEWS OF
uted to the development of a panagglutinating warm-
reactive IgG autoantibody, as no new alloantibodies MULTIPLY TRANSFUSED PATIENTS
were detected. Additional transfusions were withheld,
and the patient made a slow recovery with supportive A number of reports have described a systematic
care, even though the autoantibody persisted in the approach to the issue of the development of autoanti-
patient’s serum. Another patient was a 64-year-old bodies and AIHA following transfusion.
woman with advanced carcinoma of the breast. Castellino and colleagues147 reviewed the medical
Although her course was prolonged and complicated records of 184 pediatric patients with sickle cell disease
and included an autologous peripheral blood stem cell who had received multiple RBC transfusions. They
transplant, the authors attributed the development of found that 14 children (7.6%) developed warm (IgG)
AIHA to blood transfusion. She received multiple trans- RBC autoantibodies. Median transfusion exposure at
fusions and ultimately developed an anti-E alloanti- the time of autoantibody formation was 24 RBC units
body, a positive DAT due to IgG and complement RBC (range, 3–341 units). The autoantibody reacted as a
sensitization, and a panagglutinin in the serum and panagglutinin in 11 patients but had anti-e specificity
eluate from her RBCs. A course of corticosteroid therapy in three patients. RBC-bound complement also was
resulted in resolution of her hemolysis. detected in five patients. Clinically significant hemoly-
Szymanski and Smith143 reported five patients who sis was documented in four patients, in each of whom
developed autoantibodies following transfusion. In both RBC-bound IgG and C3 were detected. The
three of the patients, the development of autoantibo- development of RBC autoantibodies was associated
dies was attributed to transfusion of fresh-frozen with the presence of alloantibodies. The authors con-
plasma (FFP) to patients who had an alloantibody (anti- cluded that formation of warm autoantibodies in asso-
D). The authors determined that FFP contains a small ciation with transfusions is not rare in pediatric
number of RBCs prior to freezing and suggested that patients with sickle cell disease.
RBC debris in Rh-positive FFP might elicit RBC autoan- Singer and colleagues148 evaluated 64 transfused
tibodies and AIHA if patients have prior allo-anti-D. patients with thalassemia and found that 14 (22%)
Mohn and coworkers144,145 performed a unique set developed RBC alloantibodies, in part due to RBC phe-
of experiments that led to the production of autoanti- notypic differences between donors (mostly white) and
338 Immune Hemolytic Anemias

patients (75% Asian). Sixteen patients (25%) developed Thompson and colleagues158 studied a panel of 72
a positive DAT and had autoantibodies in their serum. human monoclonal antibodies (anti-A, -D, -C, -c, -E, -e,
Eleven autoantibodies were IgG panagglutinins, and -G, -Jka, -Jkb). Although these antibodies were highly
five were IgM antibodies of which two had anti-i specific for their respective blood group antigens, they
specificity. Of the 16 autoantibodies, 13 were transient were found to react with unrelated foreign antigens
and did not cause significant hemolysis. Three patients and autoantigens (DNA, thyroglobulin, hemoglobin,
developed severe WAIHA and required prolonged or rabbit and rat tissues, rheumatoid factor). IgM mono-
recurrent treatment with steroids. Two patients clonal antibodies showed cross-reactivity more often
required splenectomy. Autoimmunization was associ- than did IgG antibodies. Only five of 44 IgM antibodies
ated with alloimmunization and with the absence of (one of 13 anti-D, two of 7 anti-E, one of 13 anti-c, and
the spleen (44% and 56%, respectively). one of two anti-A) failed to demonstrate any cross-
Aygun and coworkers149 reviewed the medical reactivity. The results clearly demonstrated that a con-
records for pediatric and adult SCD patients who had siderable proportion of antibodies that are produced in
been transfused over a 10-year period. They found response to exogenous antigen can show multispecific
that 8% and 9% of pediatric and adult patients, respec- properties identical to those of natural autoantibodies.
tively, developed RBC autoantibodies. They noted The authors argued against the concept that the anti-
that neither partial nor extended RBC phenotype bodies originated from a multispecific pool of B cells
matching had been utilized. and were undergoing a transition from multispecificity
to monospecificity by the process of somatic mutation
and affinity maturation. The ability of the antibodies to
IMMUNOLOGICAL DATA make the extremely fine allotypic distinctions among
several hundred blood group antigens makes it
There have been interesting reports on lymphocyte unlikely that they are broadly reactive antibodies
populations following transfusion. Kaminski and needing a refinement of specificity by somatic muta-
associates150 showed that T-lymphocytes from multi- tion. Thompson and colleagues158 suggested that mul-
transfused patients develop cytotoxicity against auto- tispecificity is a feature of antibodies produced in
logous cells; this was not necessarily associated with response to exogenous antigens. Their conclusions
tissue damage. Paglieroni and colleagues151 showed were that autoantibodies associated with autoimmune
that after transfusions, IgM levels and the absolute disease may belong to one of three classes:
number of B cells that coexpress CD5 (i.e., a B-cell sub-
population thought to produce autoantibodies) rose 1. Antibodies that apparently are monospecific for the
to twice pretransfusion levels. Preexisting auto- autoantigen
antibodies (e.g., rheumatoid factor and antinuclear 2. Antibodies that show multispecific properties but
antibody) increased in four of five transfused patients. whose primary antigen is the autoantigen
Phage display technology and human monoclonal 3. Autoantibodies that show multispecific properties
antibody characteristics are yielding important data and whose autoreactivity is unrelated to the pri-
pertinent to the foregoing discussions. Marks and col- mary antigen (e.g., blood group antibodies)
leagues152 and Ouwehand and coworkers153 isolated
several blood group-specific antibodies (anti-B, -HI, Another interesting publication pertinent to the fore-
-D, -E, -Kpb) from an RNA-derived “naive” phage dis- going discussion is that by Barker and Elson.159 These
played V-gene repertoire. RNA was obtained from the authors demonstrated that T cells from healthy
mononuclear cells of two nonimmunized D-positive humans could be stimulated by multiple epitopes on a
individuals, one from blood group O and one from self-protein to yield primary proliferative responses in
blood group A. Three of the antibodies (anti-D, -Kpb, vitro. Synthetic 15-mer peptides, corresponding to the
and -HI) were made against antigens for which the sequence of the human Rh polypeptide associated with
donors were positive. For example, the RNA-derived Cc/Ee RBC antigens, were used to stimulate T cells
B-lymphocyte repertoire from D-positive donors con- from donors whose RBCs were C+e+. Reproducible
tained anti-D reactivity. The serologic reactivity of the responses were obtained. The proliferation could be
anti-D resembled that of a typical “cold” antibody. The blocked by antibodies to HLA-DR but not by anti-
affinity for the D antigen was low (kD 5.2 × 107M–1), as bodies to HLA-DP or -DQ. The responding cells were
was expected from a VH gene repertoire derived from most commonly derived from the CD45RA+ subpopu-
μ-positive B lymphocytes. The anti-E (derived from an lation, which indicated that they had not been activated
E-negative donor) cross-reacted with E-negative RBCs in vivo. Thus, Rh-specific T cells appeared to be
at 4°C. Ouwehand and coworkers153 felt that these data “immunologically ignorant” of a wide range of self-
were in concordance with observations of others that epitopes that they recognize. Barker and Elson159
the B-cell repertoire contains a considerable level of proposed that these peptides represent cryptic determi-
self-reactivity. They also felt that their data provided a nants that either are not normally generated by the pro-
molecular basis for the serologic cross-reactivity cessing of proteins in antigen-presenting cells (APCs)
(“mimicking”) characteristics commonly described in or that bind with low affinity to major histocompatib-
AIHA.154–157 ility complex class II molecules. They further suggested
Unusual Aspects of Acquired Immune Hemolytic Anemias 339

that Rh autoantibodies (e.g., those in AIHA) might epitope-specific immune response to subdominant epi-
result from the stimulation of immunologically topes on that protein. The specificity of the immune
ignorant Rh-specific T cells. These responses could be response spreads to include self-epitopes other than the
triggered by the presentation of previously cryptic self- one that initiated the inflammatory process. In this sce-
determinants by APCs, as a consequence of a change in nario, infection, organ transplantation, or autoimmu-
antigen processing.159 nity (organ-specific or systemic) leads to tissue
In a later publication, Barker and associates160 pro- damage. The resulting inflammation and tissue
posed that pathogenic autoantibody production in damage then primes a hierarchial cascade of autoreac-
many cases of AIHA is driven by the activation of T- tive T-cell specificities, even allowing cryptic or
helper cells specific for previously cryptic epitopes on sequestered epitopes to be processed and presented.160
the Rh proteins. Two panels of overlapping 15-mer
peptides, corresponding to the sequences of the 30-kD THE SOURCE OF AUTOANTIBODIES
Rh proteins associated with the expression of the D
and Cc/Ee blood group antigens, were synthesized FOLLOWING TRANSFUSION
and screened for the ability to stimulate the in vitro
proliferation of mononuclear cells from the peripheral The RBC autoantibodies that develop in some patients
blood or spleen of nine patients with AIHA. Culture following blood transfusion could be produced by the
conditions were chosen that favor recall proliferation immune system of the patient or by immune cells in the
by previously activated T cells rather than primary blood products that had been transfused. Definitive
responses. In seven of the patients—including all four studies have not been done to determine whether such
patients with autoantibody to the Rh proteins—two or autoantibodies have been of donor or recipient origin.
more peptides elicited proliferation, but cells from As indicated previously, some investigators have
eight of nine patients with other anemias and seven of suggested that the autoantibodies probably are pro-
nine healthy donors failed to respond to the panels. duced as a result of a breakdown of the recipient’s
Multiple peptides were also stimulatory in two posi- immunologic tolerance, possibly as a result of expo-
tive control donors who had been alloimmunized sure to closely related antigens.122–125,132 Also, auto-
with Rh D-positive RBCs. Six different profiles of pep- antibodies have been produced by transfusions of
tides elicited responses in the patients with AIHA; this noncellular blood products,144,145 thus suggesting that
variation could reflect the different human leukocyte the host immune system is the source of the autoanti-
antigen (HLA) types in the group. Stimulatory pep- bodies. In 1980, we suggested that RBC autoanti-
tides were identified throughout domains shared bodies might be developed by immune cells of the
between or specific to each of the related 30-kD Rh donor as a result of their persistence in the recipient
proteins, but T cells that responded to nonconserved following transfusion.6
regions did not cross-react with the alternative Viable lymphocytes that are capable of responding
sequences. HLA class II antibodies blocked the to phytohemagglutinin have been demonstrated in
responses, and depletion experiments confirmed that blood stored at 4°C for up to 1 month.161–163 Schechter
the proliferating mononuclear cells were T cells. and associates164 were among the first to study the
Notably, splenic T cells that proliferated against mul- possibility of the persistence of donor lymphocytes in
tiple Rh peptides also responded to intact RBCs. The recipients following blood transfusion. They noted a
researchers proposed that ignorant T cells are acti- fivefold or greater rise in atypical lymphocytes or in
vated in AIHA due to the coincidence of two inde- vitro 3H-thymidine incorporation by blood leukocytes
pendent events: persistent stimulation by (or both) occurring one week after blood transfusion.
cross-reactive environmental antigen(s) and Th1- These values declined to pretransfusion levels by the
lymphokine-induced changes in autoantigen process- third week. Lymphocytotoxic reactivity was also
ing that trigger the presentation of many previously detected in six of 12 patients. A later report by
cryptic epitopes. Thus, it is possible for healthy indi- Schechter and colleagues165 described cytogenetic
viduals to harbor activated Rh-specific T cells after studies that were performed on the peripheral blood
encountering environmental antigen(s) that cross- leukocytes of 10 adult patients who received fresh
react with the Rh proteins; however, in the absence of blood from donors of the opposite sex. They demon-
a coincidental change in autoantigen processing, the strated spontaneously dividing cells of both donor
corresponding Rh epitopes remain cryptic, and no and host origin in some patients, suggesting that their
disease results. The current findings—that T cells data indicated the counterpart of the in vitro mixed
from patients with AIHA but not from healthy indi- leukocyte reaction and that dividing donor cells might
viduals can respond to native RBC proteins processed represent a subclinical graft-vs.-host reaction.
by APCs—are consistent with this interpretation.160 Further studies by Adams,166 Houbiers,167 and
A typical immune response against a self- or foreign Lee168 and their colleagues all used sensitive poly-
protein is usually focused on one or two epitopes that merase chain reaction assays to track the survival of
are termed dominant within that protein. Epitope allogeneic donor cells following transfusion. These
spreading was initially defined as the diversification of studies showed rapid clearance of donor cells with
epitope specificity from the initial focused, dominant little or no persistence beyond 5 to 7 days. In these
340 Immune Hemolytic Anemias

reports, however, the donor-recipient pairs were been pregnant. If such diseases are indeed caused by
unmatched for HLA, and the transfused blood was allogeneic antibody-producing cells, then the term
either stored or buffy coat-depleted. Different results autoimmune is inappropriate. The question then arises
were found when fresh allogeneic blood was whether autoimmune diseases are really alloimmune
exchanged between mothers and their children. For diseases.
example, when fresh, nonirradiated maternal blood Could AIHA be caused by a similar mechanism? We
was transfused to fetuses, cells with the maternal suggested this in 1980.6 The following remarkable
karyotype have been found to circulate in the case (Patient 6), which is described by Ishikura and
newborn for up to four years.169 colleagues192 and also is reviewed in Chapter 12, sup-
Shapiro170 described a case of AIHA with the clini- ports our suggestion that autoantibodies can be pro-
cal features of runt disease in a 3-month-old child. He duced as a result of long-standing microchimerism
suggested that maternal blood might have gained following transfusion.
access to the baby’s circulation and that maternal lym-
phoid cells had survived and set up a graft-vs.-host P A T I E N T 6 : The patient received a liver transplant at
reaction. Similar cases have been described in adults age 11 months because of congenital biliary atresia. The
with acquired hypogammaglobulinemia171,172 and in blood type of both donor and recipient was group A,
children with dysgammaglobulinemia.173 Rh(D) positive. He received multiple transfusions during
Studies in a murine transfusion model indicated the surgery and postoperative course. On day 198 after
that donor WBCs can persist for more than 6 weeks the surgery, massive gastrointestinal bleeding occurred,
and can mediate immunologic function, as indicated and the DAT was positive for the first time (33 days after
by transient transfer of dinitrochlorobenzene sensi- the last blood transfusion of RBCs). The antibody eluted
tivity.174 These investigators also indicated that the from the patient’s RBCs showed panagglutination that
clearance of donor WBCs in the murine transfusion included panel cells, cord cells, Rh null, and -D- cells. Two
model is much slower than in humans. Indeed, days after transfusion of least incompatible RBCs, severe
caution should be exercised in any extrapolation to hemolysis occurred, with progressive anemia (hemoglo-
humans of conclusions drawn from results in trans- bin 4.0 g/dL), hyperbilirubinemia (direct, 16.6 mg/dL;
fused mice.175 indirect, 6.2 mg/dL), hemoglobinuria, and high LDH
Lee and coworkers176 characterized survival kinet- (2546 IU/L). The hemolysis subsided following therapy
ics of donor WBC subsets in transfused patients and with plasmapheresis, whole blood exchange, cyclophos-
found multilineage persistence of donor WBCs (CD4, phamide pulse therapy to suppress antibody production,
CD8, CD15, CD19) for 6 months to 11/2 years after and high-dose gamma globulin. The DAT became nega-
transfusion in 7 of 10 female trauma patients. They tive 31 days later. Analysis of IgG heavy-chain allotypes
suggested that studies of microchimerism are critical (Gm) in this case produced remarkable findings. Allotypes
to prevention of alloimmunization and transfusion- of the donor and recipient were identical prior to trans-
induced graft-vs.-host disease and, potentially, to plantation; however, Gm typing of the eluate from the
induction of tolerance for transplantation. patient’s RBCs revealed numerous allotypes and indicated
There is a growing body of evidence that microchi- that the antibodies could have been produced by neither
merism, which might develop as a result of fetoma- the liver donor nor the patient.
ternal cell trafficking during pregnancy and delivery,
could be involved in a number of autoimmune The authors concluded that the antibodies were
diseases such as systemic lupus erythematosus derived from two or more persons other than the
(SLE), scleroderma, and juvenile dermatomyosi- donor and recipient. Therefore, they suggested that
tis.177–191 Microchimerism can occur in the child the observed delayed hemolytic anemia might have
and/or in the mother. been caused by alloantibodies produced by donor
Fetal cells have been shown to persist in maternal lymphocytes derived from the patient’s multiple
peripheral blood for decades after childbirth , and it has transfusions.
been suggested that SLE and scleroderma could be The foregoing concept192 supports our suggestion
forms of chronic graft-vs.-host reaction caused by fetal from 19806 and is consistent with observations made
or maternal cells that have crossed the placenta and by others of patients with delayed HTRs in which a
have remained unrecognized by the host due to histo- positive DAT has persisted for a longer period of time
compatibility.177,180–184 Indeed, Johnson and associ- than the transfused cells could have survived (see
ates182 demonstrated male cells in every histologically Chapter 14).193,194
abnormal tissue type that was examined in a multi- Dzik195 has proposed that microchimerism be
parous woman who died of complications of SLE, but viewed as occupying a middle position on a spectrum
such cells were not found in histologically normal between alloimmunization and graft-vs.-host disease.
tissue. It has further been suggested that autoimmune Between these extremes, there could be circumstances
diseases might occur as a result of microchimerism, in which donor cells are neither quickly eliminated
which could originate from a blood transfusion, from a from the host nor allowed unchecked clonal expan-
twin, or from the mother. Such a diversity of possibili- sion. Instead, a balanced state of microchimerism
ties is applicable to men and to women who have never might ensue.
Unusual Aspects of Acquired Immune Hemolytic Anemias 341

C. Autoimmune Hemolytic Anemia in Infancy


and Childhood

Although AIHA in infancy and childhood is similar in


TABLE 9-8. CLINICAL FEATURES AT INITIAL
many ways to AIHA in adults, some distinctive
PRESENTATION OF 25 CHILDREN WITH
findings have been emphasized in reviews on this
AUTOIMMUNE HEMOLYTIC ANEMIA
topic. Compared with AIHA in adults, the disorder in
children more often appears acutely with a violent
Acute* Chronic
onset but a good prognosis; there is a more frequent Clinical Features (N = 9) (N = 16)
association with acute viral infections; underlying
chronic disorders are less common; and PCH is much Age (mean) 5.6 years 4.7 years
more common.196-202 Sex
Female 6 8
Male 3 8
Associated disease 1 8
CLINICAL FINDINGS AND COURSE Viral prodrome 7 10
Onset of symptoms
Typically, in AIHA of infancy and childhood there is a Abrupt† 8 8
Chronic 1 8
sudden onset of pallor and malaise, often following an Presenting symptoms
intercurrent infection. Slight jaundice, splenomegaly, Pallor 9 14
and hepatomegaly are found in about half of the sub- Jaundice 6 14
jects (Table 9-8).199 Lethargy 5 10
Dark urine 5 4
Most authors indicate that a higher percentage of Physical exam
children have acute, self-limited disease compared Hepatomegaly 5 11
with adults. This tendency was particularly striking in Splenomegaly 4 14
the series reported by Buchanan and colleagues,198 in Lymphadenopathy 4 2
which this course was recorded in 17 of 22 children * Cure by six months.
(77%). The authors commented that in this respect, † Onset of symptoms less than one week.

AIHA in childhood resembles idiopathic thrombocy- From Heisel MA, Ortega JA: Factors influencing prognosis in childhood
autoimmune hemolytic anemia. Am J Pediatr Hematol Oncol 1983;5:147–152.
topenic purpura, which is also usually acute and tran-
sient in childhood but chronic in adults.
This is also true of the patients described in the Zupanska and coworkers196 reported on 44 children
excellent review of 80 children with AIHA followed in with AIHA; in more than 50% of patients, the disorder
one department by Habibi and associates.197 They was preceded by an acute infection or immunization,
reported that, in their experience, 42.5% of children and these cases were associated with acute or suba-
and only 4.5% of adults had transient hemolysis cute forms of the disease. In contrast, patients with an
(duration less than 3 months). They pointed out clini- accompanying disease generally developed chronic
cal and immunologic features that tended to distin- anemia.
guish transient cases from chronic cases (Table 9-9). Sokol and associates199 reported that most hemolytic
Onset of AIHA at an older age and in association with episodes followed an acute infection, which frequently
an underlying disorder (other than an acute infection) was mild and often involved the upper respiratory
was associated more commonly with chronic disease. tract. The infection tended to be current or very recent.

TABLE 9-9. COMPARISON OF CLINICAL FEATURES IN CHILDREN WITH TRANSIENT OR CHRONIC AUTOIMMUNE
HEMOLYTIC ANEMIA

Transient Cases Chronic Cases

Onset in first 4 years of life Frequent (80%) Moderate incidence (44%)


Clinical presentation Invariably acute or hyperacute Acute in only 34% of cases
Hemoglobinuria 45% 4%
Prodromal acute infection 70% 4%
Underlying disorders 5.8% 58%
Effectiveness of corticosteroid therapy Rapid and constant Variable
Clinical course Full recovery in less than 3 months Intermittent or permanent course for months or years
Direct antiglobulin test Only complement detected on RBC in 67.6% IgG or IgG with complement in 84.7% of cases
of cases
Mortality None 11.2%

From Habibi B, Homberg JC, Schaison G, Salmon C: Autoimmune hemolytic anemia in children. A review of 80 cases. Am J Med 1974;56:61–69.
342 Immune Hemolytic Anemias

Sometimes, a brief period of apparent recovery pre-


ceded the onset of hemolysis. Only two of 42 children TABLE 9-10. AIHA IN CHILDHOOD AND
in this series were found to have an underlying chronic ADOLESCENCE: DISTRIBUTION OF CASES
disorder. Diagnoses included WAIHA, cold agglutinin Number of Patients
syndrome, PCH, and mixed AIHA (Table 9-10).
Cold AIHA
The series reported by Gurgey and colleagues203 is
unusual in that the disorder was acute and transient in Mixed
Age and Sex WAIHA PCH CAS AIHA
only 18 of 51 children, whereas 33 children had chronic
disease. Similarly, Heisel and Ortega204 found that nine 0–4 years
of 25 patients had acute disease, whereas 16 patients Male 7 10 2 1
had chronic disease. All of the patients who were diag- Female 4 4 0 0
nosed at 12 years of age or older and eight of 10 5–9 years
Male 1 1 1 0
patients who presented at less than 2 years of age were Female 2 1 1 1
determined to have chronic disease (Fig. 9-5). 10–15 years
Most patients have idiopathic AIHA, although the Male 2 0 0 2
disorder has been associated with a number of Female 0 1 1* 0
underlying diseases, 197,205 including immune * Autoantibody of IgG class.
deficiency disorders,205 autoimmune diseases such Modified from Sokol RJ, Hewitt S, Stamps BK, Hitchen PA: Autoimmune
as systemic lupus erythematosus and other collagen haemolysis in childhood and adolescence. Acta Haematol 1984;72:254–257.

diseases,196,204,206,207 Hodgkin’s disease,204,208 serum


IgA deficiency, and giant cell hepatitis.210,211 10
Acute AIHA
Chronic AIHA
LABORATORY FINDINGS 8

Anemia in AIHA of childhood is often


Number of children

severe198,199,204,205,212 (Table 9-11); hemoglobin levels 6


were below 6.0 g/dL in 28 of the 42 children reported by
Sokol and coworkers.199 The rate of fall of hemoglobin
was quite spectacular in some patients (e.g., 3 g/dL in
4
24 hours in one patient and 1.8 g/dL in 12 hours in
another).199 Reticulocytopenia contributes to the seri-
ousness of the anemia in some patients.199,204,213,214 For a
more complete discussion of reticulocytopenia in AIHA, 2
see the section on reticulocytopenia in Chapter 3.
Platelet counts are normal in the majority of
patients; on occasion, high values are seen during 0
periods of active hemolysis.199 Thrombocytopenia is <2 2–12 > 12
present in 14% to 32% of patients, however, and in Age at diagnosis (years)
some instances is severe and contributes to morbidity
FIGURE 9-5. Distribution of 25 patients according to age at diagnosis.
and mortality.196-198,204,205,212,215 (From Heisel MA, Ortega JA: Factors influencing prognosis in childhood
Serological findings most commonly are those of autoimmune hemolytic anemia. Am J Pediatr Hematol Oncol
WAIHA, and CAS is reported only uncommonly.196,197 1983;5:147–152.)

TABLE 9-11. LABORATORY DATA AT TIME OF DIAGNOSIS

Laboratory Data Acute (N = 9) Chronic (N = 16)

Hemoglobin (mean) 4.3 g/dL (Range 2.7–7.6 g/dL) 5.3 g/dL (Range 2.7–9.4 g/dL)
Reticulocyte count (mean) 9.4% (Range 1.9–31.5%) 34.6% (Range 0.1–79.7%)†
Nucleated red blood cell present 4 11
White blood cell count (mean) 12,600/mm3 (Range 4,700–30,300/mm3) 18,900/mm3 (Range 7,000–37,000/mm3)
Early white blood cell present 2 7
Platelet count (mean) 421,000/mm3 (Range 191,000–583,000/mm3) 195,000/mm3 (Range 15,000–795,000/mm3)‡
Thrombocytopenia (<150,000/mm3) 0 10*

* p < 0.01.
† p < 0.02.
‡ p < 0.05.
From Heisel MA, Ortega JA: Factors influencing prognosis in childhood autoimmune hemolytic anemia. Am J Pediatr Hematol Oncol 1983;5:147–152.
Unusual Aspects of Acquired Immune Hemolytic Anemias 343

PCH is much more commonly reported among chil- was positive at birth and remained positive in 1977 and
dren than among adults; in some series, it makes up a 1978, and evidences of hemolysis were still present. The
significant percentage of patients.200-202 Indeed, Sokol RBC count was 2.2–2.3 million/mm3, reticulocytes
and colleagues199 reported that a Donath-Landsteiner were 13.8%–23.4%, and spherocytes were found on the
antibody was found in 17 of 42 of their patients (40%) peripheral blood film. The autoantibody was of
(see Table 9-10). the warm type and possibly of anti-e specificity. The
A number of cases with unusual serological child was treated with corticosteroids and responded
findings have been reported. For example, Friedmann well; the DAT became negative, only to become
and coworkers216 reported a 9-year-old girl with positive again when corticosteroids were reduced.
Evans’s syndrome who had high-titer, high thermal Splenectomy at age 3.5 years resulted in significant
amplitude (37°C) complete IgM autoantibody. Despite improvement, although evidence of hemolysis was still
aggressive management, the patient ultimately died present in 1989.
of complications of her disease. Such a clinical course
is consistent with the dismal prognosis among adults Management
with similar warm-reactive IgM autoantibodies.217-220
Issitt and colleagues109 described a case of WAIHA in Habibi and colleagues197 reported that corticosteroid
which marked depression of RBC Rh antigen expres- therapy seemed to be effective frequently, but it was
sion resulted in the patient presenting with severe often difficult to exclude the possibility of sponta-
anemia but a negative DAT. Unlike two previously neous remission, especially in acute cases. Patients
reported cases cited by these researchers, in which the who respond to steroids frequently demonstrate a
diagnosis of AIHA was established before the DAT significant decrease in hemolysis within 10 days of
became negative, the authors’ patient presented with diagnosis. Splenectomy was performed for 16 patients
negative serological findings during his first episode who showed corticosteroid resistance or dependence
of anemia. Win and associates221 reported an infant with severe side effects. Eight patients were fully
with AIHA who had an autoantibody with anti-Kpb cured of hemolysis, although positive serologic
specificity. findings remained in three of these; moderate
Hemoglobinuria is much more common in children improvement occurred in four patients, and four
than in adults, which is due partly to the more fre- others were not evaluable. The efficacy of immuno-
quent occurrence of PCH. Hemoglobinuria occurred suppressive agents was estimated in seven patients
in all 17 patients reported by Sokol and associates199 who had been given regular and sufficient doses for
who had Donath-Landsteiner antibodies, but it also periods of longer than 4 months. Four patients were
occurred in patients with cold agglutinins and in judged to have benefited with development of com-
mixed-type AIHA. (PCH is discussed in more detail in pensated hemolysis and reduced corticosteroid
Chapter 5.) dosage.
Sokol and associates199 stated that corticosteroids
and blood transfusion were the mainstays of treat-
AIHA OCCURRING AT A VERY EARLY AGE ment and that splenectomy was performed on only
four patients who had hemolysis that was not con-
Sokol and associates199 reported that their youngest trolled by the initial therapy. “Spectacular responses”
patient was 3 months old. Other reports, however, occurred in three of the four cases, but in the fourth
include patients with AIHA occurring at even earlier patient (who also had thrombocytopenia), hemolysis
ages,197,204,212,222,223 including one patient who was recurred 1 year later. Similarly, Buchanan and col-
diagnosed at the age of 2 weeks.224 Erler and cowork- leagues198 reported beneficial effects among three of
ers225 described an infant who was found to have a four patients who underwent a splenectomy.
positive DAT at birth without evidence of maternal On the other hand, Heisel and Ortega204 and
alloantibodies or autoantibodies. No antibody was Johnson and Abildgaard227 reported poor results with
found in the infant’s serum, and he had no evidence splenectomy, which was used as a therapy for patients
of hemolysis, but an eluate from his RBCs contained with chronic AIHA not responsive to corticosteroids.
an IgG panagglutinin. The DAT was still positive Five patients underwent splenectomy, and in no case
6 weeks after birth but was negative at 6 months of was this treatment associated with complete resolu-
age. The authors suggested that autoantibody pro- tion of the disease.204 Three of these patients died
duction had occurred in utero. within 1 year, all with evidence of sepsis (see Chapter
Hadnagy226 provided a brief report of a child whose 11 for a detailed discussion of overwhelming post-
AIHA dated from birth and had continued for 13 years splenectomy infection, or OPSI).
at the time of publication. The child was born severely Olcay and coworkers213 described a 4-month-old
jaundiced in 1976 and was exchange-transfused on the patient with AIHA and reticulocytopenia who did not
third day after birth. There was no evidence of fetoma- respond to corticosteroid therapy of 7 days’ duration
ternal incompatibility, and there were no unexpected but was successfully treated when a combination of
antibodies in the mother’s serum. The infant’s DAT immunosuppressive therapies (cyclophosphamide, 6-
344 Immune Hemolytic Anemias

mercaotppurine, and IVIG) were added to the cortico- 6 mg/kg/day, additional RBC transfusions were
steroids. In contrast, Heisel and Ortega204 reported poor administered, and 36 hours after admission, the
results for seven patients with chronic disease who were authors began plasma exchange. They initially used
steroid dependent and were treated with immunosup- plasma as the replacement, but prior to completion of
pressive therapy that included azathioprine, cyclophos- the exchange, one unit of RBCs was infused instead
phamide, 6-mercaptopurine, hydrochloroquine sulfate, of plasma. The patient remained clinically stable
and chlorambucil. Only one complete remission was during the next 2 days and after a final transfusion
observed among these patients. Duru and associates209 was discharged on oral prednisone with a hematocrit
treated two patients with cyclosporine without benefit. of 32% and a reticulocyte count of 1.5%. After an
Anecdotal case reports have indicated success using additional week of treatment, prednisone was dis-
high-dose intravenous immunoglobulin therapy. continued, and there was no recurrence of hemolysis
Sasaki and coworkers228 reported a 7-week-old male or detectable antibody after 1 year of follow-up. The
infant with WAIHA who had normal levels of serum authors suggested that plasma exchange could be
IgG, IgA, and IgM and normal concentrations of indicated for selected patients with fulminant AIHA
whole complement, C3, and C4. He was treated with who cannot be stabilized with steroid and transfusion
RBC transfusions and prednisolone (3 mg/kg/day) or therapy alone.
methylprednisolone (1.5 mg/kg/day). After 15
weeks, his hemoglobin was less than 5 g/dL, and
during the therapy he had pneumonia, a subcuta- PROGNOSIS
neous abscess, and sepsis. On day 110, he began a
course of IVIG at a dose of 0.4 g/kg for 5 days. At that There is a wide range of mortality rates among
time, corticosteroid therapy was switched from various reported series of cases. Most authors indicate
methylprednisolone to betamethasone. The hemoglo- that many cases are acute and transient, but that there
bin level improved rapidly, and the DAT became neg- is a significant mortality rate among patients with
ative. Six months later, he was well, the hemoglobin chronic illness, especially if this related to an underly-
was 13.0 g/dL, reticulocytes were 5%, and the DAT ing disorder.
was negative even though betamethasone had gradu- In the series of Habibi and coworkers,197 80% of the
ally been withdrawn. patients had acute, transient disease with no deaths,
Otheo and colleagues229 reported a 4-year-old child but there was an 11.2% mortality among chronic cases.
with Evans’s syndrome who presented with a hemo- These patients generally had associated disorders and
globin level of 5.2 g/dL, reticulocytes 20%, and inherent complications at the time of death: thrombo-
platelets 55,000/μL. The DAT was positive with anti- cytopenia and cerebral hemorrhage, malignant vari-
IgG but negative with anticomplement serum. A non- cella, septicemia, cytomegalovirus infection, marrow
specific IgG RBC autoantibody was demonstrated, as aplasia, and immune deficiency disease.
were antiplatelet antibodies. Initial treatment Poschmann and Fischer231 reported that the mortal-
included IVIG at a dose of 800 mg/kg/day for 5 days ity rate of patients with AIHA appears to be consider-
and transfusions of RBCs. The hemoglobin and ably lower among children than among adults. In
platelet counts rapidly reached normal values, and their review of the literature, they found that the
the reticulocyte count was 2.7% 5 weeks after IVIG. mortality among children was 10%–28%, whereas in
The patient was given maintenance doses of IVIG (800 adults, it was 28%–70%.
mg/kg) every 2 weeks for 8 months, although there Zupanska and associates196 stated that, although
was no longer any trace of autoantibody by the fifth the prognosis in AIHA is generally good, about
month. Eighteen months later, the patient still had 10% of their children died. In no case, however, was
normal hematological values. death due directly to severe anemia. The children
The successful use of plasma exchange has also died from uncontrollable bleeding caused by throm-
been reported. McConnell and colleagues230 described bocytopenia, uremia, or the accompanying disease
the course of a boy aged 2 years and 10 months, who (e.g., SLE).
was admitted with a hemoglobin level of 5.1 g/dL, a Carapella de Luca and colleagues205 reported that
reticulocyte count of 0.7%; hemoglobin in the urine in eight of their 29 patients (28%) died—two as a direct
the absence of RBCs, DAT positive for C3, IAT posi- consequence of acute hemolysis. These two deaths
tive for a Donath-Landsteiner antibody (anti-P), and occurred 1 and 20 days, respectively, after diagnosis of
a cold reactive antibody with anti-IH specificity. Bone AIHA was made.
marrow aspiration revealed marked erythroid Sokol and coworkers199 found that recovery was
hypoplasia and phagocytosis of RBCs by macrophages. rapid and that complete recovery occurred in 83% of
He was treated with 2 mg/kg/day of prednisone and their 42 patients, usually within 6 months. Two
transfusions, but his hemoglobin concentration fell patients died—one from profound hemolysis shortly
progressively to a nadir of 2.9 g/dL, hemoglobinuria after admission and the other of miliary tuberculosis.
worsened during transfusions, and the reticulocyto- Heisel and Ortega204 reported a mortality rate of
penia persisted. The steroid dose was increased to 25% among their 16 patients with chronic AIHA.
Unusual Aspects of Acquired Immune Hemolytic Anemias 345

Pregnant
Delivery Pregnant Thrombophlebitis
? Pulmonary
Splenectomy embolus Delivery Delivery

(mg. per day)


Corticoid
200

100

FIGURE 9-6. Clinical course of idio- 40


pathic AIHA in 25-year-old female

Reticulocytes (percent)
through repeated pregnancies. Hematocrit

Hematocrit (percent)
(From Swisher SN: Acquired 30
hemolytic disease. Postgrad Med
1966;40:378–386.)

20

10
Reticulocytes

June Sept. Dec. 1953 1954 1955 1956 1957 1958 1959
1952

D. Autoimmune Hemolytic Anemia During


Pregnancy

Because AIHA commonly occurs in young women, it is Serologic findings are those of WAIHA in a large
not surprising that there are reports of AIHA during majority of patients, although some reports have
pregnancy. Coincidence alone, however, does not fully included patients with cold antibodies234,238 or with
explain the coexistence of these two conditions, as both warm and cold antibodies.237,239
pregnancy has definite effects on the course of AIHA.
Swisher232 described a patient with idiopathic AIHA,
whose degree of hemolysis was increased with each of MATERNAL FINDINGS
three pregnancies and remitted with each parturition
(Fig. 9-6). Similarly, Benraad and associates233 de- Chaplin and associates237 reviewed reports of 19
scribed a woman who developed AIHA during her first pregnancies with presumed AIHA. All patients had
pregnancy; during her second pregnancy, AIHA devel- unequivocal evidence of acquired hemolysis, but evi-
oped once again. Also, Sokol and colleagues234 found dence of an autoimmune etiology was often incom-
that AIHA was at least four times more frequent during plete. The DAT on maternal cells was positive in seven
pregnancy as in the nonpregnant female population, patients, negative in five, and not performed in seven.
and concluded that it is most unlikely that the associa- Four of the five mothers with a negative DAT were
tion of AIHA with pregnancy is a mere coincidence. treated with corticosteroids, and all improved.
Hoppe and coworkers235 reported an increase in The hazard to maternal survival was often ex-
autoimmunization against RBCs during pregnancy, tremely serious. In nine patients, the hemoglobin fell
and Buyon236 pointed out that there is also a significant below 5 g/dL, and in another eight instances, hemo-
effect of pregnancy on other autoimmune diseases. globin values were in the range of 5–8 g/dL.
The clinical findings vary widely among patients, Leukopenia was present in four pregnancies and
with most women responding well to therapy and thrombocytopenia in three. Vigorous transfusion
delivering a fetus with minimal or no evidence of therapy, high doses of corticosteroids, and/or early
hemolysis. Anemia in some patients can be so induction of labor were employed as life-saving
severe as to threaten the patient’s life, however, and measures for critically ill patients.
maternal red cell autosensitization also can induce Particularly significant is the observation that
fetal hemolysis, requiring management as for hemolysis worsened as the pregnancy progressed in
hemolytic disease of the newborn (see Chapter 13). 18 of the 19 patients. Complete or partial remissions of
Accordingly, close observation throughout preg- hemolysis occurred in 16 patients within 3 months
nancy and appropriate therapy are critical to a suc- after delivery. The two patients who did not improve
cessful outcome.234,237 subsequently were diagnosed as having SLE.
346 Immune Hemolytic Anemias

Sokol and colleagues234 reported much less serious


clinical findings for 20 pregnant patients with RBC OUTCOME OF PREGNANCIES
autoantibodies. The researchers indicated that auto-
sensitization occurred in one in 50,000 pregnancies, Four of the 19 pregnancies reviewed by Chaplin and
although only seven of the patients in their report had coworkers237 resulted in the delivery of a stillborn pre-
hemolytic anemia. The serological abnormalities in all mature infant, and a fifth premature infant died of
cases were discovered during pregnancy. Most patients bronchopneumonia 48 hours after birth. No hemato-
had warm autoantibodies, although three had cold logic data were reported on these five infants, and none
antibodies, and both warm and cold antibodies were were described as having pallor, jaundice, or hydrops.
found in one patient. The clinical presentation varied All of the infants, however, were born to mothers with
from severe hemolytic anemia (lowest hemoglobin severe anemia (hemoglobin <5 g/dL), which suggests
between 5 g/dL and 8 g/dL) to merely finding sero- that fetal death was related more to the critical mater-
logical abnormalities in otherwise well patients. nal state than to autoantibody-induced hemolytic
Treatment of AIHA with corticosteroids was necessary disease of the newborn. Accordingly, the authors advo-
in only three patients in whom the hemolysis was cated a policy of frequent adjustment of corticosteroid
severe. Eight of the patients had 11 further pregnancies, dosage during pregnancy in an effort to maintain the
and all remained asymptomatic, although some had patient’s hemoglobin above 10 g/dL.
weak red cell autoantibodies and one patient had active Among the four patients in group one in the report of
disease. The authors concluded that the risks associ- Issaragrisil and Kreuatrachue,240 there were three spon-
ated with AIHA occurring in pregnancy were less than taneous deliveries (with normal infants in two of the
thought previously provided that the mother received cases) and one stillbirth. The fourth patient, who had
treatment promptly. SLE, developed toxemia of pregnancy, and her hemo-
The severity of clinical findings among the patients globin level declined from 7.1 g/dL to 3.7 g/dL within
reported on by Issaragrisil and Kruatrachue240 are 5 days in spite of receiving prednisolone, 60 mg/day.
intermediate between the findings described by Labor was therefore induced for this patient. The
Chaplin and colleagues237 and Sokol and cowork- mother delivered a low-birth-weight infant; both
ers.234 They described 14 cases, which they divided mother and child survived.
into two groups. Group one consisted of four Of six patients in group two who developed hemo-
patients who were diagnosed during pregnancy, and globin values below or equal to 6.7 g/dL, spontaneous
group two consisted of 10 patients who became preg- abortion occurred in three patients, and therapeutic
nant during the remission period of previously diag- abortion was performed for two patients because of a
nosed AIHA. lack of response to prednisolone therapy. One patient
All four patients in group one were first found to had a spontaneous delivery. After termination of the
have AIHA when they were in the third trimester of pregnancies, the mothers’ hemoglobin levels responded
pregnancy—at term in two cases and in the seventh well to glucocorticoid therapy.
month in the other two. The two cases that were not Only five pregnancies in this group continued into
diagnosed until term had been seen earlier in preg- the third trimester. Anemia in two patients responded
nancy and found to have normal hemoglobin levels at well to prednisolone (40 mg/day), and one patient
that time. Three patients were treated with cortico- did not need corticosteroid therapy. These pregnan-
steroids without improvement in their hemoglobin cies resulted in spontaneous deliveries with normal
values which, prior to delivery, ranged from 3.7 g/dL infants. A cesarean section was performed in one case
to 7.4 g/dL. After deliveries, three patients responded due to severe preeclampsia and fetal distress; both
well to cortocosteroid therapy, but one patient with mother and child survived. One patient, who had SLE
SLE had hemoglobin levels that waxed and waned and nephrotic syndrome, had a spontaneous abortion
between 6.5 g/dL and 8.0 g/dL. followed by acute renal failure and later died.
In contrast to the patients in group one, seven of It should be noted that corticosteroid therapy for
10 patients in group two presented during the first the patients reported on by Issaragrisil and
trimester of pregnancy with a falling hemoglobin, Kruatrachue240 generally was started only after
including three cases in which anemia was significant anemia occurred. Patients were not treated
detected during the first month. Except for the at a time when obvious hemolytic anemia was present
anemia, the first trimester was uneventful, although if the hemoglobin was in the range of 9.0 g/dL.
one patient had a therapeutic abortion for reasons Indeed, some patients were not treated with corticos-
unrelated to hemolysis. A second phase of anemia teroids at hemoglobin values 6.7–7.4 g/dL. Although
occurred more abruptly somewhere between the such hemoglobin values are generally well tolerated,
third and fourth months of gestation, however. perhaps more favorable outcomes would have
Severe anemia developed in three patients (hemoglo- occurred if an attempt had been made to cause remis-
bin 4.0–4.7 g/dL), and moderate anemia occurred in sions in the patients’ hemolysis before the sharp drops
four patients (hemoglobin 6.7–7.9 g/dL). In two in hemoglobin occurred later in their pregnancies.
other patients, the lowest hemoglobin values were This is in keeping with the suggestions of Chaplin and
9.7 g/dL and 11.7 g/dL. colleauges237 and Sokol and coworkers.234
Unusual Aspects of Acquired Immune Hemolytic Anemias 347

NEONATAL HEMOLYSIS infant’s hemoglobin was 15.8 g/dL and the DAT and
IAT were negative. Nevertheless, at 2 months, the
Hemolytic disease of the newborn, usually mild but hemoglobin had dropped to 8.6 g/dL, and there were
occasionally requiring therapy, has been reported spherocytes along with anisocytosis, poikilocytosis,
among infants born to mothers with AIHA. In some but and broken cells on the peripheral smear. Although no
not all instances, there is documentation of an immune antibodies were demonstrated in the infant’s serum or
pathogenesis of the hemolysis caused by the maternal on his RBCs, the authors attributed the drop in hemo-
autoantibody traversing the placenta. Lawe241 reported globin to placental transfer of maternal autoantibodies.
on a 25-year-old woman with apparently inactive
Hodgkin’s disease who developed fulminant AIHA in
the 37th week of pregnancy. The baby required four INFANTS WITHOUT HEMOLYSIS
exchange transfusions for hyperbilirubinemia. The
same IgG antibody was found in mother and infant. In In spite of the rather significant complications of
the case reported by Bauman and Rubin,242 the mother pregnancy involving the mothers in the series
had a strongly positive DAT, and both her serum and reported by Issaragrisil and Kruatrachue,240 there
an eluate from her RBCs contained a broadly reactive was no evidence of hemolysis in any of the infants. In
autoantibody. The newborn had a positive DAT and a the review of Chaplin and colleagues,237 hematologi-
compensated hemolytic process in the first week of life. cal characterization of the infants was inadequate for
The infant’s hemoglobin fell to a low of 9.7 g/dL, but evaluation in 16 of the 19 pregnancies. Nevertheless,
no specific therapy was required. Vedovini and 11 infants were inferred as normal because no hema-
Benedetti243 reported a case of AIHA that developed tologic abnormalities were noted, even though the
during the last month of pregnancy and resulted in mothers had “brisk hemolysis” during the latter
hemolysis in the newborn, which was treated with months of pregnancy. Sacks and coworkers250 also
steroids and blood transfusions. The mother did not point out that severe maternal hemolytic anemia does
receive steroids until after delivery. In the report of not necessarily correlate with neonatal outcome.
Sokol and associates234 concerning seven patients with These authors reported a patient with a positive DAT
AIHA, three infants were mildly affected with and IgG autoantibody in her serum who nonetheless
hemolytic disease due to the maternal autoantibodies maintained a hemoglobin above 13 g/dL without
crossing the placenta, but no treatment was needed. treatment. The neonate also had a positive DAT and
In other reports, however, the immune etiology of IAT but no evidence of hemolytic disease.
the newborn’s hemolysis is not clear. For example, In addition, a number of other case reports of well-
Burt and Prichard244 described an infant who required documented AIHA in pregnancy indicate that the
two exchange transfusions during the first 26 hours of newborns were healthy with no evidence of hemoly-
life to control hyperbilirubinemia. Both the mother sis.233,248,251-254 In all of these cases—including one case
and infant had negative DATs and IATs; however, the of Evans’s syndrome248 and one report of WAIHA in
authors suggested that the diagnosis could have been two successive pregnancies—the mothers were
WAIHA because of the clinical setting and because treated successfully with corticosteroids during preg-
there was improvement in the hemolytic process in nancy.233
the mother following splenectomy. The blood type of
both mother and infant was group A, Rh positive, and
there was no evidence of maternal alloimunization. HEMOLYTIC ANEMIA OF PREGNANCY
Soderhjelm245 reported an infant who became WITH A NEGATIVE DIRECT
severely anemic and jaundiced in the fourth week of
life and seemed to respond to therapy with cortico-
ANTIGLOBULIN TEST AND FREQUENT
steroids. Seip246 described an infant who was severely RECURRENCES
anemic (hemoglobin 2.5 g/dL; reticulocytes 22%) and
leukopenic (WBC 1800/μL) at 2 months of age and In addition to the cases already cited, there are other
required multiple transfusions. The mother, who had reports of hemolytic anemia in pregnancy in which
SLE, also had leukopenia during pregnancy. Although the DAT is negative and the cause of the hemolysis is
the anemia in both infants was attributed to placen- uncertain.251,255-260
tally transferred IgG autoantibody, the infants had Kumar and associates260 reported on a patient who
negative or equivocal DATs. developed severe life-threatening hemolytic anemia
A number of cases of Evans’s syndrome in preg- during the last trimester of three successive pregnan-
nancy have been reported.247,249 Passi and associates249 cies, with spontaneous recovery 11–12 weeks post-
reported on a patient who presented with a hemoglo- partum after each pregnancy. She remained normal
bin of 7.7 g/dL and a platelet count of 36,000/μL. After during the entire nongravid state. Extensive investi-
treatment with corticosteroids, her hemoglobin gations were performed to find the cause of the
improved to 10.2 g/dL, and the platelets varied hemolysis, but none was documented. In particular,
between 60,000 and 80,000/μL. She delivered a 2.6-kg the DAT was negative; there were no RBC antibodies;
male infant at 37 weeks of gestation, and at birth, the tests for congenital hemolytic anemias were negative
348 Immune Hemolytic Anemias

in the patient and her family; the blood film was TABLE 9-12. COMPLEMENT FIXING ANTIGLOBULIN
normal; and no RBC membrane abnormalities could CONSUMPTION TEST IN A PATIENT WITH
be found. Corticosteroids and high-dose IVIG were RELAPSING HEMOLYTIC ANEMIA OF PREGNANCY
ineffective, and she was treated with blood transfu- AND A NEGATIVE DIRECT ANTIGLOBULIN TEST
sions. The authors commented that this was the only
example of its kind that came to their attention among Molecules of IgG/RBC
55,000 deliveries conducted over the course of a
decade in their hospital. Prepartum Postpartum
Starksen and colleagues258 reviewed cases of unex-
plained hemolytic anemia during 19 pregnancies in Mother 212 53
Newborn — 250 (cord blood)
12 women. They described striking anemia beginning <25 (14 days postpartum)
in the first (38%) or second (38%) trimester, resolving
within 5 months after delivery (18 of 19 patients, 95%) From Yam P, Wilkinson L, Petz LD, Garratty G: Studies on hemolytic
and recurring in a subsequent pregnancy (six of 12 anemia in pregnancy with evidence for autoimmunization in a patient
with a negative direct antiglobulin (Coombs’) test. Am J Hematol
patients [50%]). Corticosteroid treatment, when 1980;8:23–29.
employed, has resulted in a uniformly favorable
response.251,256,258 35
Transient hemolysis was present in four of 19 (22%)
infants. In a number of instances, detailed hema-
tologic evaluations have been carried out, which

Hematocrit (%)
30
excluded folate deficiency, hemoglobinopathies, red
blood cell enzymopathies, hereditary spherocytosis,

on Prednisone
PNH, PCH, and SLE.251,257,258 An extracorpuscular

Delivery
abnormality seems to be present, as a number of case 25
reports document that transfused RBCs did not
survive normally.256,261,262
Yam and colleagues251 studied a patient with 20
hemolytic anemia of pregnancy with a negative DAT
10
and applied the CFAC test in an effort to document
Reticulocytes (%)

an immune pathogenesis (Patient 7) (Fig. 9-7). 8

P A T I E N T 7 : The patient was a 34-year-old woman 6


who had had seven previous pregnancies. She had a 4
single blood count during her seventh pregnancy; the
hematocrit level was 29% and reticulocytes were 6.7%; a 2
single hematocrit level of 39% was available from her
0
records in the non-gravid state. On referral, during the 1 2 3 4 5 6 7 8
second trimester of her eighth pregnancy, her hemoglobin Months
level was 9.6 g/dL, the hematocrit level 28% and the
reticulocyte count 10.3%. She had a hemolytic anemia as FIGURE 9-7. The clinical course of a patient with relapsing hemolytic
anemia of pregnancy with a negative direct antiglobulin test. Hemolytic
manifested by persistent anemia (hematocrit 24–27%) anemia was evident prior to the initiation of therapy with prednisone,
and reticulocytosis (6.3–10.3%) (Fig. 9-7). as manifested by anemia with reticulocytosis. The anemia resolved, but
The patient’s DAT was negative throughout her preg- reticulocytosis persisted during prednisone therapy until the delivery of
nancy using polyspecific antiglobulin serum and multiple the patient’s baby. Thereafter, the reticulocytosis subsided and pred-
nisone therapy was gradually withdrawn (see text). (From Yam P,
dilutions of monospecific anti-IgG, -C3, and -C4, and no
Wilkinson L, Petz LD, Garratty G: Studies on hemolytic anemia in preg-
antibody was detected in her serum or in an eluate from her nancy with evidence for autoimmunization in a patient with a negative
red cells. Hereditary spherocytosis was suspected because direct antiglobulin (Coombs’) test. Am J Hematol 1980;8:23–29.)
her blood film revealed a moderate number of spherocytes,
but six of her seven children were examined, and none
showed evidence of spherocytes. The peripheral blood tum revealed a significant reduction in RBC sensitization. At
films, hematocrits, and reticulocyte counts were normal in all that time, only 53 molecules of IgG per RBC were present.
six children, thus making hereditary spherocytosis unlikely. The baby was anemic at birth, with a hemoglobin of 12.2
A CFAC test performed on the patient’s blood revealed g/dL. The CFAC test on a cord blood sample revealed
212 molecules of IgG per red cell (normal <25) (Table 9- 250 molecules of IgG per RBC. The infant did not require
12). The patient was treated with 50 mg of prednisone therapy, and 14 days later only 25 molecules of IgG per
daily during the last 12 weeks of her pregnancy, which RBC were detectable (Table 9-12).
caused an increase in her hematocrit from 24% to 27% to
31% with a reticulocyte count of 7.5% (Fig. 9-7). After par- Starksen and colleagues258 reported on a patient
turition, the prednisone was gradually decreased and then whose CFAC test during pregnancy revealed a result at
discontinued. The CFAC test performed 2 weeks postpar- the upper limit of normal (40 molecules IgG per red
Unusual Aspects of Acquired Immune Hemolytic Anemias 349

blood cell, with an abnormal range reported by that of AIHA and for patients with hemolytic anemia with a
laboratory as >40 molecules of IgG per RBC). Other negative DAT. There are no reports of autoantibody-
techniques used to document a possible immune mech- mediated hemolytic disease of the newborn requiring
anism for the hemolysis were a five-times concentrated exchange transfusion in mothers whose AIHA is well
ether eluate reacted against enzyme treated RBCs and controlled with corticosteroids during pregnancy.
an enzyme-linked antiglobulin test for cell-bound IgG. Although the incidence of hemolytic disease of the
The results of both these tests were negative. newborn requiring therapy is low, amniocentesis
Kumar and coworkers260 summarized reported cases appears indicated in the presence of brisk hemolysis,
of DAT-negative hemolytic anemia of pregnancy as which is mediated by an IgG autoantibody.237,251 Some
follows: In most cases, the hemolytic anemia occurs authors have reported early termination of pregnancy
during the last trimester of pregnancy, although in in instances in which corticosteroids were not effective
some cases hemolysis is noted in the first and second in controlling hemolysis. In contrast to alloantibody-
trimesters. Most commonly, its first occurrence is noted induced hemolytic disease of the newborn, early termi-
in the primigravida state, but instances of hemolysis nation of pregnancy has been primarily for unrelenting
first occurring in the fifth and seventh pregnancies maternal anemia but would be reasonable if amniocen-
have been reported. Most patients show spontaneous tesis indicates high fetal risk (see Chapter 13).
reversal to normalcy within 2 months postpartum, For patients who do not respond to corticosteroids
although hemolysis can persist up to the fifth month and who have brisk hemolysis with severe anemia,
after delivery. The majority of patients have a single additional or alternative therapeutic measures should
recurrence in a subsequent pregnancy, but relapsing be considered.
hemolysis has been reported in as many as three to five
successive pregnancies. The anemia is usually severe
and even life threatening. Most often, it requires THE EFFECTS OF IMMUNOSUPPRESSIVE
support with blood transfusions. Bjornson and associ- DRUGS DURING PREGNANCY
ates259 had to transfuse their patient with a total of 121
units of blood for relapsing hemolytic anemia in five Although the use of corticosteroids and other immuno-
successive pregnancies. Corticosteroids and IVIG have suppressive drugs during pregnancy causes significant
been reported to be of value but not confirmed to be concerns, there is an extensive medical literature
universally effective. The majority of infants born to regarding the use of these drugs during pregnancy in
mothers with this type of hemolytic anemia show evi- other autoimmune diseases (Table 9-13).264,265 Immuno-
dence of transient hemolysis lasting for 1 to 2 months. suppressive drugs other than corticosteroids seem not
Although extensive investigations have not docu- to have been used for treatment of AIHA in pregnant
mented the cause of hemolysis, several findings patients, but they could be reasonable therapeutic alter-
suggest the possibility of an immune mechanism. natives when the risk of severe anemia unresponsive to
There are no documented intracorpuscular defects, corticosteroids appears to be greater than the risk of the
transfused RBCs have a shortened survival time,256,262 use of immunosuppressive drugs.
corticosteroids and IVIG are often effective, and
hemolysis occurs in some neonates.259,263
Corticosteroids
Controlled studies comparing corticosteroid-treated
MANAGEMENT and nontreated patients during pregnancy have failed
to show an increased incidence of birth defects,266 and a
The potential seriousness of AIHA in pregnancy, par- National Institutes of Health consensus opinion indi-
ticularly in inadequately treated patients, justifies cates that antenatal corticosteroid therapy reduces mor-
very close observation throughout pregnancy of any bidity in preterm infants with little or no effect on
patient known to have a history of AIHA or who is long-term physical or psychologic development and
discovered to have AIHA while pregnant. Antenatal only a questionable increase in perinatal infection in
hematologic evaluation appears indicated every two both the mother and her neonate.267 Accordingly, corti-
weeks until the eighth month of pregnancy and costeroids are the most widely used immunosuppres-
weekly thereafter until delivery. Such evaluations sive drugs in pregnancy and are probably the safest.
should include as a minimum a blood count and a Nevertheless, significant risks include premature
reticulocyte count. Changes in the strength of the DAT rupture of the membranes, intrauterine growth retarda-
and IAT might offer additional information, although tion, and accentuation of several pregnancy-induced
there no data are available on the correlation of maternal complications, including hypertension, gesta-
autoantibody titers and the severity of autoantibody- tional diabetes, osteoporosis, and osteonecrosis.264
induced hemolytic disease of the newborn.
Patients with evident hemolysis should be treated
with corticosteroid therapy in doses adequate to main-
Azathioprine
tain a hemoglobin level above 10 g/dL. This is true Most large-scale studies of transplant patients have
both for patients with characteristic serological findings demonstrated that azathioprine is fairly well tolerated
350 Immune Hemolytic Anemias

TABLE 9-13. SUMMARY OF TOXICITIES OF IMMUNOSUPPRESSIVE AGENTS IN ANIMALS AND HUMANS

Animals Humans

Drug Embryotoxic Fetal Maternal Fetal Breast-feeding

Glucocorticoids No Cleft palate Premature rupture of Small for gestational-age Crosses into breast
Aggressive behavior membranes offspring milk at low
Hypertension Adrenal hypoplasia concentration,
Glucose intolerance Promotes lung maturity well tolerated
Osteoporosis Cleft palate*
Osteonecrosis Stillbirth*
Chlorambucil Yes Small for gestational-age † Small for gestational-age Contraindicated*
offspring offspring
Skeletal abnormalities Skeletal abnormalities
Renal agenesis Renal agenesis
Cyclophosphamide Yes Small for gestational-age Decreased fertility in Small for gestational-age Contraindicated
offspring males and females offspring
Skeletal abnormalities Limb abnormalities
Cleft palate Coronary artery
Exophthalmos agenesis
Decreased fertility Late onset tumors in
offspring*
Azathioprine Yes Skeletal abnormalities † Diminished fertility in Contraindicated‡
Cleft palate offspring*
Decreased thymic
development and
hematopoiesis
6-mercaptopurine Yes Cleft palate † Small for gestational-age †
offspring
Prematurity
Intrauterine growth
retardation
Cleft palate
Methotrexate Yes Skeletal abnormalities † Embryotoxic Contraindicated‡
Cleft palate Skeletal abnormalities
Facial abnormalities
Cyclosporin A Yes Renal tubular cell Renal insufficiency Spontaneous abortions Contraindicated
damage Decreased lymphocyte
subsets
Intravenous † † † Small for gestational-age †
immunoglobulin offspring
Autoantibodies

* Theoretical risk, or case reports only.


† No information available.
‡ Conflicting information.
From Bermas BL, Hill JA: Effects of immunosuppressive drugs during pregnancy. Arthritis Rheum 1995;38:1722–1732. Reprinted by permission of Wiley-Liss, Inc., a
subsidiary of John Wiley & Sons, Inc.

during pregnancy. There have been isolated reports of


Cyclosporine
immunoglobulin deficiency, chromosomal abnormali- Cyclosporine’s lack of bone marrow toxicity and its low
ties, and malformations, but it is not clear whether the carcinogenic potential make this medication an appeal-
rate of these events is significantly greater than that ing alternative for patients who do not respond to more
seen in a healthy obstetric population. Another unan- conventional immunosuppressive therapies.264 There
swered question is whether treatment of mothers with are increasing data on offspring exposed to cyclospo-
azathioprine during pregnancy will affect the germ-cell rine in utero because more and more female transplant
lines of offspring and therefore affect the future fertility recipients survive to childbearing age. In one series, 154
of those offspring. In cases where immunosuppression pregnancies were evaluated in renal transplant reci-
is necessary during pregnancy, however (e.g., trans- pients treated with cyclosporine in addition to other
plant recipients, severe autoimmune disease), Bermans immunosuppressive agents (prednisone and azathio-
and Hill264 suggest that azathioprine could be a reason- prine).268 Overall, complications among newborns
able drug of choice. Janssen and Genta265 are more cau- were slightly lower than complications observed
tious and state that the drug should be reserved for among patients treated with other immunosuppressive
pregnant women whose (rheumatic) diseases are agents, and no malformations were seen. Other investi-
severe or life threatening. gators, however, reported a high rate of spontaneous
Unusual Aspects of Acquired Immune Hemolytic Anemias 351

abortion and preterm labor in women treated with


cyclosporine and prednisone during pregnancy.269 TABLE 9-14. PROPOSED TREATMENT
Renal function in children has been reported to be RECOMMENDATIONS DURING PREGNANCY
normal.270 Whether there will be long-term immuno- FOR PATIENTS WITH AUTOIMMUNE DISEASE*
modulatory effects on offspring who have been
Moderate-Severe
exposed to cyclosporine in utero is unknown. Mild Disease Disease Life-threatening
Bermas and Hill264 suggest that cyclosporine can be
used successfully in pregnancy, whereas Janssen and No treatment Steroids Steroids—high dose
Genta265 state that, in general, the drug is contraindi- Steroids—low dose Cyclosporin A Cyclosporine
cated during pregnancy. Azathioprine Azathioprine
Cyclophosphamide†

Other Drugs * Methotrexate. chlorambucil. and 6-mercaptopurine should be avoided.


† Life-threatening illness with no alternative treatment.

Drugs such as chlorambucil, cyclophosphamide, 6- Modified from Bermas BL, Hill JA: Effects of immunosuppressive drugs during
pregnancy. Arthritis Rheum 1995;38:1722–1732. Reprinted by permission of
mercaptopurine, and methotrexate should generally be Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
avoided during pregnancy.264 High-dose intravenous
immunoglobulin (IVIG) could be a reasonable treat-
ment, although it is not entirely benign, as there have
been case reports of hemolytic disease of the newborn for this indication during pregnancy. Only rarely will
severe enough to require exchange transfusion after the splenectomy become necessary, but consideration
mother received IVIG during pregnancy.271 should be given to this option if it appears that mater-
nal benefits outweigh the increased risk for fetal death.
Conclusions Several authors, in discussing therapy of idiopathic
thrombocytopenic purpura (ITP), have advised against
One must keep in mind that much of the literature on splenectomy in pregnancy, citing a maternal mortality
the use of immunosuppressive drugs during pregnancy rate of 10% and a high neonatal mortality rate due to
has been developed from reports of patients who have premature labor.272-274 Nevertheless, splenectomy has
received a transplant, and in these cases, immunosup- been performed successfully during pregnancy for
pressive therapy is essential. These drugs, other than patients with ITP and for thrombocytopenia associated
corticosteroids, will be necessary in only very unusual with the antiphospholipid syndrome.275-279 The tech-
instances in the management of AIHA. Bermas and nique of laparoscopic splenectomy has been used by
Hill264 provide treatment recommendations during some.278,279 Gottlieb and colleagues275 state that for
pregnancy for patients with autoimmune disease and patients with ITP, splenectomy during pregnancy
transplant recipients (Table 9-14). The evidence suggests should be carried out on maternal indication if there is
that corticosteroids, azathiporine, cyclosporine, and poor or no response to medical treatment, or if medical
IVIG might be tolerated, but methotrexate, chlorambu- treatment is hazardous to the mother. They further state
cil, 6-mercaptopruine, and cyclosporine should be that strong warnings against surgical treatment during
avoided. pregnancy should be revised because of improvements
in surgical technique and care of the premature infant.
For patients who develop severe AIHA that is
THE ROLE OF SPLENECTOMY poorly responsive to corticosteroids during preg-
nancy, an elective postpartum splenectomy should be
Although splenectomy is an effective measure for considered as a means of preventing recurrent prob-
AIHA, we are not aware of it having been performed lems in a subsequent pregnancy.251

E. Familial Autoimmune Hemolytic Anemia

There is a rather significant body of medical literature Even more common are reports of family members
regarding the occurrence of AIHA in more than one of patients with AIHA who have been afflicted with
member of a family (Table 9-15).280-304 Among reported another type of autoimmune disease. There are many
cases of familial AIHA are four sets of twins, of which reports of patients with AIHA in whose family one or
three sets were reported as being identical.293,300,305,306 more members suffer from an immunologically medi-
On the other hand, Dacie263 followed a patient for ated disorder other than AIHA.263 Pirofsky289,290
20 years after the diagnosis of AIHA had been made, reported that one or more relatives of eight of 43 (19%)
and during this time her twin remained free from the of his patients had suffered from a wide range of
disease. McLeod and associates303 reported the clinical probable or possible autoimmune disorders, including
course of three siblings, all of whom had Evans’s syn- glomerulonephritis, pernicious anemia, Stevens-
drome in childhood. Johnson syndrome, rheumatoid arthritis, SLE, and
352 Immune Hemolytic Anemias

TABLE 9-15. OCCURRENCES OF AIHA IN MORE thrombocytopenic purpura. Conley307 found an even
THAN ONE MEMBER OF A FAMILY higher incidence of autoimmune disorders among
family members: 14 of 33 patients (42%) had family
Relationship and Age at Onset members known to have an immunologically mediated
(Years, Except Where Stated) Reference disorder similar to those listed by Pirofsky or from a
lymphoma or leukemia. Conley and Savarese298
Mother (61) and daughter (23) 280 reviewed the occurrence of other autoimmune and
Mother (72) and son (33) 281, 282
Two sisters (18 and 14) 283
lymphoproliferative disorders among patients with
Sister (55) and brother (38); a further sister AIHA and emphasized the frequent association of
(35) possibly affected 284 abnormal serological reactions and abnormalities in the
Two children had “symptoms of acquired levels of serum immunoglobulins.
hemolytic anemia” “Two brothers The development of AIHA and the finding of other
developed autoimmune hemolytic anemia” 285
Mother (40) and daughter (8) 286 manifestations of autoimmunity among family mem-
Sister (57) and brother (who had died earlier bers of numerous patients suggests that a fortuitous
of AIHA and lymphosarcoma) 287 relationship is unlikely. It is more probable that
Sister (3 months) and two brothers (3 and genetic factors play an etiologic role. The nature of the
4 months) 288
Two sisters (66 and 49) and brother (48) 289, 290
genetic mechanism, however, is unknown. Conley
Two sisters (4 and 12) 291 and Saverese298 suggested that an abnormality of cells
Brother (14) and sister (19); also paternal aunt 292 of the immune system, often genetically determined,
Identical twin girls (1 and 21⁄2) 293 could predispose to serologic changes, immune defi-
Mother and daughter (12) 294 ciency, autoimmune diseases, and neoplasia.
Sister (40) and brother (62) 295
Brother (20) and two sisters (17 and 15) 296
Two sisters (66 and 62) 297
Two sisters (68 and 60) 298
Mother and son (14) 299
Twin brothers (21 and 23) 300
Two sisters (71 and 73) 301
Two families with two affected sisters (7 months
and 6, and 12 months and 9 months) 302
Three siblings with Evans’s Syndrome 303

Modified from Conley CL, Savarese DM: Biologic false-positive serologic tests
for syphilis and other serologic abnormalities in autoimmune hemolytic anemia
and thrombocytopenic purpura. Medicine (Baltimore) 1989;68:67–84.

F. Cardiac Surgery and Cold Autoantibodies

Development of hemolysis in patients with cold agglu- a low of 12.6%. The cold agglutinin was an anti-I with
tinins of high thermal amplitude, or exacerbation of the a titer of 51,200 against adult (I) RBC at 4°C in albumin
rate of hemolysis in patients with cold agglutinin syn- and a titer of 100 at 30°C.
drome (CAS), are risks of cardiac surgery under condi- Wertlake and colleagues314 attributed hemolysis
tions of hypothermia. There is also the potential for that began during cardiac surgery under hypothermia
intracoronary hemagglutination with inadequate distri- to an anti-HI cold agglutinin that destroyed A2B
bution of cardioplegic solutions, thrombosis, embolism, donor RBCs in an A1B patient. Preoperatively, the
ischemia, or infarction.308-311 When measures have been hematocrit was 45%, but it dropped progressively to a
taken to avoid the hematologic and cardiac conse- level of 30% on the fifth day after surgery and did not
quences of exposure to cold in patients with cold agglu- begin to rise until day 15 despite a reticulocytosis. The
tinins, the reported results have been excellent.312 cause of the hemolysis was somewhat uncertain,
however, because the direct Coombs’ test was consis-
tently negative, and the thermal amplitude of the anti-
ADVERSE EVENTS body was elevated only transiently to a level of 22°C
on the sixth day after surgery.
Although complications are not common, there are Izzat and coworkers311 reported a case in which
well-documented events precipitated by cold during agglutination of RBCs occurred within 1 minute of ini-
surgery among patients with cold agglutinins of high tiation of antegrade cold blood cardioplegia at 10°C
thermal amplitude. Bedrosian and Simel313 reported a and led to embolization in the coronary microcircula-
patient with well-compensated chronic CAS who had tion. After the multiple agglutinates were noted, a
an acute “life-threatening” hemolytic crisis during an coronary sinus cannula was inserted through the right
elective herniorrhaphy in a cool (20°C) operating atrium, and continuous retrograde cold crystalloid
room. The patient’s hematocrit decreased from 36% to cardioplegia was infused. Agglutinates were noted to
Unusual Aspects of Acquired Immune Hemolytic Anemias 353

45 Rectal temperature
Prefusate temperature
Hemagglutination
40

35
Critical
temperature 29⬚

Temperature (⬚ C)
30
FIGURE 9-8. Clinical determination of
critical temperature for hemagglutina-
tion (29°C) (CPB, cardiopulmonary 25
bypass). (From Diaz JH, Cooper ES,
Ochsner JL: Cardiac surgery in patients
with cold autoimmune diseases. Anesth 20
Analg 1984;63:349–352.)

15

0 15 30 45 60 75 90
Induction Cooling Cardioplegic Rewarming Discontinue
on CPB infusion CPB

Time (minutes)

flush back from the coronary arteries into the aortic surgery, the heart was arrested using a 4°C blood car-
root. Nevertheless, the patient did not show any signs dioplegia solution, and agglutinated blood was found
of cardiac damage postoperatively. when the anterior descending artery was incised.
Diaz and associates315 observed large RBC aggluti- Postcardioplegia reperfusion of the heart was begun
nates within the coronary vessels and the coronary with a blood cardioplegia solution delivered at 37°C
sinus when a patient’s rectal temperature decreased under controlled conditions of flow and pressure.
from 36°C to 30°C during cardiopulmonary bypass. Postoperatively, there was no grossly evident hemolysis
This led to the diagnosis of previously unsuspected and no evidence for myocardial necrosis. The patient’s
CAS. They stated that the most important principle in convalescence was uneventful. It should be noted that
the immediate management of unanticipated cold the patient’s cold agglutinin was an anti-I that had an
hemagglutination is the early determination and agglutination titer of 256 at 4°C and a titer of 4 at 24°C
maintenance of body temperature above that highest but did not react at 30°C or 37°C. The fact that the cold
temperature (critical temperature) at which the cold agglutinin was of low thermal amplitude might have
antibody reacts (Fig. 9-8). mitigated against the development of complications.
Bracken and colleagues316 described cardiopul- Stalker318 reported that clumps of agglutinated RBCs
monary bypass performed on two patients who had can plug the microcirculation and cause myocardial
cold agglutinins that had gone undetected prior to necrosis, and Davidsohn and associates319 reported
surgery. In one patient, a 67-year-old male, the red hepatic infarcts in mice injected with anti-RBC serum.
cells in the cardioplegia heat exchanger were clumped Shahian and colleagues308 stated that cold-mediated
and separated from the plasma, and the patient devel- RBC agglutination within the coronary microcircula-
oped hemoglobinuria. On the evening of surgery, the tion might lead to inadequate distribution of cardio-
patient was noted to have a cold, pulseless left leg and plegic solution, poor myocardial protection, and
underwent a bedside revascularization procedure. He perioperative infarction. They cautioned that unless
died of hemodynamic compromise on the second day cold antibodies are identified prior to surgery or by
after surgery. The authors commented that it is not the surgeon or perfusionist during bypass, such
clear whether cold agglutinins were directly related to sequelae of microscopic RBC agglutinates may be
the terminal event. attributable to other causes. Indeed, the numerous
Holman and coworkers317 reported on a patient who other causes of hemolysis during cardiopulmonary
developed intracoronary agglutination of the blood car- bypass, including mechanical trauma, will likely be
dioplegia solution. The patient was a 69-year-old male considered before testing for the presence of cold
with severe pulmonary dysfunction and class IV angina agglutinins with a high thermal amplitude.320
pectoris who was referred for bypass grafting of a prox- Although not related to surgery, other instances of
imal stenosis of the anterior descending artery. During exacerbation of CAS with exposure to cold have been
354 Immune Hemolytic Anemias

reported. Niejadlik and Lozner321 described a patient occurs at room temperature. Antibodies that are reac-
who developed acute, severe hemolysis after being tive at temperatures of 30°C or higher are capable of
placed on a cooling mattress. The patient was a 51- causing hemolysis (see Chapter 5), and special precau-
year-old white female who was hospitalized with fever tions would seem to be warranted during hypother-
and right lower lobe pneumonia. Her hematocrit on mia, even though the manifestations of hemolysis
admission was 44%, but after being placed on a cooling might be minimal to none prior to exposure to cold.
mattress, signs of intravascular hemolysis developed In some cases, autoagglutination will first be
with marked hemoglobinuria, and her hematocrit noticed in the operating room.311,315-317,323 Indeed,
decreased to a low of 16%. Mycoplasma pneumoniae Dake and coworkers323 and Bracken and colleagues316
infection was documented, and cold agglutinins were have recommended that observation of the blood car-
present at a titer of 25 at 4°C, a titer of 32 at room tem- dioplegia system for agglutination before cardioplegia
perature, and a titer of 8 at 37°C. administration and systemic cooling be a routine part
Colmers and Snavely322 reported on a patient with of the perfusion checklist.
primary atypical pneumonia and cold agglutinin syn-
drome with a cold agglutinin titer of 1280 whose hemo- PATIENTS WITH COLD AGGLUTININS
globin decreased from 7.5 g/dL to 4.5 g/dL overnight
following three general body sponges using iced WHO DO NOT APPEAR TO BE AT RISK
alcohol. The drop in hemoglobin was accompanied by
passage of dark-brown urine. After recovery from her Normal persons may have cold agglutinins that react
pneumonia and hemolysis, the authors performed an at 4°C with a titer of 32 or less and with a thermal
experiment in which the patient’s right arm was amplitude of less than 20°C. Although cold agglu-
immersed for 15 minutes in ice-cold water containing tinins reactive up to room temperature (~20°C) might
pieces of ice. A blood specimen was drawn immedi- seem to be of potential significance during hypother-
ately before immersion of the arm, then 15 minutes mic surgical procedures, there is an impressive lack of
after cessation of immersion and at 15-minute intervals reports of complications caused by such antibodies,
thereafter. The control and the 120-minute serums were even though they are quite common.324 Indeed, there
completely free from any trace of hemoglobin on gross is general agreement that patients with low-titer, low-
visual inspection. The 30-minute specimen was thermal amplitude antibodies can undergo operation
markedly hemolyzed, and the 45-minute specimen without any change in routine management plan.325
showed somewhat less marked hemolysis, as deter- The thermal amplitude of the cold agglutinin is of par-
mined visually and by spectral analysis. The patient ticular significance as indicated by the data of Bracken
did not develop hemoglobinuria, a fact which suggests and colleagues,316 who reported on 19 patients who
that the systemic hemoglobinemia did not reach a had cold agglutinins with titers of 64–512 at 4°C; the
sufficient concentration to appear in the urine. researchers found that only three of these patients had
cold agglutinins that reacted at 25°C. Moore and asso-
ciates326 stated that patients with titers of 32 or lower
IDENTIFICATION OF PATIENTS AT RISK at 4°C and no detectable agglutination at 28°C or
lower tolerated hypothermia well. Moore, however,
Because cold agglutinins can be detected in the sera of studied only five patients who had strongly positive
almost all individuals, the critical problem is to decide agglutination in the cold, and only one had an anti-
which characteristics of a given patient’s cold agglutinin body that reacted at temperatures as high as 23°C.
would warrant special precautions during surgery. This Nydegger and colleagues327 reported on a 51-year-
question cannot be answered precisely, but special pre- old male who needed a liver transplant and who had a
cautions would seem important when a patient has a cold agglutinin titer of 32 at 4°C, reacting with undi-
cold antibody-induced AIHA. Although cold-reactive luted plasma at temperatures up to 30°C. The authors
antibodies are not routinely characterized in detail in were concerned because the orthotopic liver transplant
the blood transfusion service, if the antibodies have a still is cool from preservation when placed into the
high enough thermal amplitude to cause AIHA, they recipient. No intra- or perioperative complications
will ordinarily be noticed during routine compatibility occurred, however.
test procedures. Also, for patients who have hemolytic AuBuchon and associates320 studied 16 patients
anemia, one would hope that a diagnosis of CAS would undergoing hypothermic extracorporeal circulation.
have been made prior to surgery on the basis of clinical They found that total extracorporeal circulation time
and laboratory findings (see Chapters 2, 5, and 6). had a significant correlation with a rise in hemoglobin
For patients without hemolytic anemia, the thermal during the procedure, but no correlation was noted
amplitude of the antibody (the highest temperature at between the extent of hemolysis and the presence of
which agglutination occurs) is probably the best guide cold-reacting autoantibodies that were present in 12
to possible complications during surgery. The thermal patients at temperatures of 22°C or below.
amplitude can be determined in the hospital’s transfu- That patients with abnormal cold agglutinin titers
sion service (see Chapter 6), and this procedure should requiring special management in hypothermic surgery
be carried out whenever strong autoagglutination are unusual is indicated by the data of Bracken and
Unusual Aspects of Acquired Immune Hemolytic Anemias 355

TABLE 9-16. TITER AND THERMAL AMPLITUDE OF to surgery in some patients.311,315,316,323 A plethora of
COLD AGGLUTININS IN 19 ADULT PATIENTS HAVING published accounts (including several reviews) have
CARDIOPULMONARY BYPASS SURGERY recommended various approaches to management of
patients who require cardiac surgery and who have
Patient Highest Temperature cold autoantibodies.308,309,311,313,315-317,323,325,326,328-345
No. Age Sex Titer Agglutination Observed* Reported techniques of cardioplegic management in-
clude warm ischemic arrest (simple aortic cross-
1 51 F 64 4°C clamping), warm blood cardioplegia, and the use of a
2 54 M 512 4°C
3 67 M 256 + at 4°C, minimal at 25°
combination of warm and cold crystalloid cardiople-
4 62 F 128 4°C gia.345 In general, cold blood cardioplegia is not appro-
5 73 F 256 4°C priate, as one would expect significant agglutination in
6 77 M 256 4°C the coronary circulation. The alternative is to use crys-
7 59 M 128 4°C talloid hypothermic cardioplegic protection. To avoid
8 77 F 64 4°C
9 65 M 512 4°C the exposure of blood to hypothermic solution in the
10 42 F 64 4°C coronary system, warm crystalloid cardioplegia can be
11 69 F 128 4°C used to flush the coronary circulation clear of blood; this
12 53 F 128 4°C is then followed by cold crystalloid cardioplegia to
13 58 M 512 4°C
14 58 M 128 4°C
induce myocardial hypothermia. Toward the end of the
15 23 M 64 4°C procedure, the heart may be rewarmed by warm crys-
16 77 F 256 25°C talloid cardioplegia before the cross-clamp is removed.
17 76 M 64 4°C More recently, continuous warm blood cardiople-
18 53 M 64 25°C gia,346–349 which can be delivered antegradely337,350,351 or
19 77 F 64 4°C
retrogradely,339–341 has been employed. This procedure
* Agglutination test performed only at 4°C, 25°C, and 37°C obviates the need for hypothermia and is recommended
From Bracken CA, Gurkowski MA, Naples JJ, et al: Case 6—1993. by a number of authors in this setting.325,339,343,352
Cardiopulmonary bypass in two patients with previously undetected cold
agglutinins. J Cardiothorac Vasc Anesth 1993;7:743–749.
THE ROLE OF PLASMA EXCHANGE
TABLE 9-17. RESULTS OF COLD AGGLUTININ IN PREPARATION FOR SURGERY
TITERS IN ADULT PATIENTS HAVING
CARDIOPULMONARY BYPASS SURGERY Plasma exchange has been used as adjunctive therapy
for some patients.309,328,340,342 This procedure requires
No. of Patients special techniques, however, as it must be performed at
temperatures above the cold agglutinin’s thermal
Surgeries requiring CPB 969 amplitude to avoid in vitro cold agglutination.328,336,353
Preoperative CA titers performed 504
Patients with titers ≥ 64 19 Also, the procedure is not reliably effective340,354,355 (see
Patients with agglutination at >4°C 3 also Chapter 11).
Change in management of case 4 Klein and colleauges328 used plasma exchange before
Change in management clearly of benefit 1 surgery for a patient with an anti-I cold agglutinin. The
Complications related to CA 1
antibody reacted strongly with RBCs at 15°C, 22°C, and
Modified from Bracken CA, Gurkowski MA, Naples JJ, et al: Case 6—1993. after papain treatment at 37°C; in addition, it aggluti-
Cardiopulmonary bypass in two patients with previously undetected cold nated several panel RBCs in saline at 30°C and occa-
agglutinins. J Cardiothorac Vasc Anesth 1993;7:743–749.
sionally reacted with saline-suspended RBCs at 37°C.
The titer at 22°C was reduced by plasma exchange from
4 to 1. During surgery, body temperature was lowered
colleagues.316 They reviewed their data regarding with the core temperature of 29°C as measured with an
adult patients seen over a 1-year period. Only 19 0f 504 esophageal sensor. There were no adverse effects
patients had a titer of 64 or greater (Table 9-16), and of during or after hypothermia. These authors point out
these, agglutination occurred at 25°C in only three that they could not be sure that difficulties would have
patients (tests were preformed at 4°C, 25°C, and 37°C developed had plasma exchange not been performed.
only) (Table 9-17). Beebe and coworkers340 described a case in which
plasma exchange was only minimally effective. Begin-
ning 10 days before surgery, the patient received
MANAGEMENT plasmapheresis every other day for five total treat-
ments, with each treatment exchanging 1.25 plasma
Hematologists, transfusion medicine specialists, and volumes with an equal volume of 5% albumin. The
cardiovascular surgeons who identify patients at risk cold agglutinin titer decreased from 4096 to 512 the day
should collaborate to develop a protocol for manage- before surgery, but on the morning of surgery, the cold
ment. It is prudent to do this in advance of need, espe- agglutinin had rebounded to a titer of 1024. The patient
cially as cold agglutinins might not be detected prior had chronic CAS but was managed throughout surgery
356 Immune Hemolytic Anemias

for splenectomy and cholecystectomy by intraopera- during a 72-minute normothermic cardiopulmonary


tive forced air convection warming; esophageal and bypass at 36°C–37°C. No ventricular fibrillation occur-
peripheral temperatures were maintained above 37°C red, and no hemolysis, hemagglutination, myocardial
throughout surgery, and she did not have an exacerba- infarction, or organ damage resulted.
tion of her hemolysis. Zoppi and associates342 reported on a 53-year-old
In the case reported by Park and Weiss,309 the patient male with abnormally elevated cold agglutinins who
had a cold agglutinin titer of 10,000 at 4°C and 5000 at was treated by two one-volume plasma exchanges.
20°C but did not have evidence of hemolysis. Plasma- There was a reduction of the titer of the cold agglu-
pheresis was performed 1 day before surgery and pro- tinins against group O RBC at 4°C from 128 to 32, but
duced “an eightfold reduction in titers.” Body the titers of 8 at 30°C and of 2 at 37°C were not
temperature was maintained above 35°C throughout affected. The surgery was successful, and the patient
coronary artery bypass grafting, which was performed was discharged on the 16th day after surgery.

G. Differentiating Delayed Hemolytic Transfusion


Reactions from Autoimmune Hemolytic Anemia

Delayed hemolytic transfusion reactions (DHTRs) are a quently rose to a peak of 24%. The blood film showed
recognized risk of blood transfusion. The reaction is small agglutinates, many spherocytes, and some myelo-
caused by the reappearance of an antibody that pre- cytes and normoblasts. The serum bilirubin was
sumably was first stimulated by pregnancy or a pre- 4.5 mg/dL, and serum haptoglobin was absent. The
vious transfusion. Unlike immediate transfusion DAT was strongly positive. The patient’s serum reacted
reactions, which are usually caused by human error, with her own RBCs and with samples from more than
delayed reactions are usually not avoidable.356-361 30 donors and thus appeared to have a nonspecific
Because hemolysis is delayed in onset (typically 3 to 14 autoantibody; however, careful serologic studies docu-
days after transfusion), the relationship of hemolytic mented the presence of anti-Fya, anti-Ce, and anti-e, and
anemia to prior transfusion might not be suspected, the patient’s hemolytic anemia resolved without
and a diagnosis of AIHA might seem more appropriate. therapy. The DAT rapidly weakened and became nega-
As in any patient with the acute onset of hemolysis, tive 10 days after the last transfusion, but the IAT
findings can include fever, pallor, jaundice, hemoglo- remained strongly positive throughout hospitalization.
binemia, hemoglobinuria, and, rarely, disseminated
intravascular coagulation or renal failure.356,362-364
Further, laboratory tests are likely to reveal the pres- DIAGNOSTIC AIDS
ence of a positive DAT and IAT, spherocytosis, and
reticulocytosis. If multiple alloantibodies or an alloan- As mentioned in Chapters 5 and 6, a presumptive diag-
tibody against a high-incidence antigen is formed, the nosis of WAIHA may be entertained for a patient who
findings can be difficult to differentiate from AIHA. has not been transfused during the previous 3 months,
The diagnostic problem is compounded by the fact has an acquired hemolytic anemia with a positive DAT,
that, in some cases, AIHA might actually develop as a does not have a cold agglutinin of high thermal ampli-
consequence of blood transfusion (see the segment of tude, has not been taking drugs known to cause
this chapter regarding AIHA following transfusion). immune-mediated hemolysis (see Chapter 8), and has
Indeed, therapy with corticosteroids for suspected a warm antibody with broad reactivity in the serum or
AIHA has been instituted365,366 or contemplated in RBC eluate. Such a presumptive diagnosis tends to be
some reported cases of DHTRs before the correct diag- correct because if donor RBCs no longer remain in the
nosis was made.364,367 patient’s circulation, alloantibodies cannot be causing
A characteristic example of a patient who had a the acquired immune hemolytic anemia. One should
DHTR simulating AIHA was reported by Croucher and note, however, that a DAT can persist for at least 3 to
colleagues.365 They described a patient with vaginal 4 months following a DHTR (see Chapter 14). Even if
bleeding who was transfused with 10 units of blood the patient has not been transfused over the previous
over a period of 8 days. Despite the arrest of hemor- 3 months, we recommend further tests to characterize
rhage, her hemoglobin level continued to fall. The the patient’s RBC antibody(ies) to avoid diagnostic
patient was found to be pale and jaundiced; her urine error and to have results of specificity tests available
was dark and contained “urobilin” but did not contain should transfusion become necessary.
hemoglobin or bile. The spleen was just palpable. Her If a patient has been transfused within recent
hemoglobin, which had been 8.5 g/dL at the time that weeks, such additional testing is mandatory to distin-
hemorrhage ceased, dropped to about 6 g/dL. At that guish between a DHTR and AIHA. The following
time, her reticulocyte count was 13%, and it subse- measures will afford important clues.
Unusual Aspects of Acquired Immune Hemolytic Anemias 357

Comparison of DAT and IAT the warm autoadsorption test, or by using the allo-
geneic adsorption test (see Chapter 10).
A simple observation that could yield valuable infor- A further clue to the differentiation of autoantibodies
mation is the comparison of the strength of the DAT and alloantibodies having Rh specificity is the
and IAT. As is stressed in Chapter 10, the DAT is specificity itself. That is, anti-e is a rare cause of a DHTR,
almost always stronger than the IAT in WAIHA. It but it is the most frequently described autoantibody
appears that autoantibody is largely adsorbed onto specificity.362 In contrast, anti-E is the most common Rh
the patient’s RBCs, and only when the RBCs are alloantibody responsible for DHTRs, but it is a relatively
heavily coated does one find a large amount of anti- unusual autoantibody.362,368 Again, if pretransfusion
body in the serum. In contrast, strongly reactive allo- RBCs are still available, it is possible to use them to dis-
antibodies might be present in a patient’s serum, but tinguish alloantibody from autoantibody with certainty.
this finding cannot result in a strongly positive DAT If the antibody shows a defined specificity, the
unless large numbers of transfused RBCs of appropri- patient’s RBCs should be tested for the relevant
ate antigenic type are present. Thus, the presence of a antigen. If it is an autoantibody, the patient’s RBCs
weakly positive DAT in association with a strongly should possess the antigen. It is not always easy to
positive IAT is presumptive evidence for the presence determine the patient’s phenotype, as DAT+ RBCs are
of an alloantibody. These findings are therefore highly difficult to phenotype and because transfused RBCs
suggestive of a DHTR. Further, even if the DAT is may be present. Chapter 6 describes some methods that
strongly positive at the time of initial evaluation, it help in phenotyping DAT+ RBCs. If transfused RBCs
can soon become weaker in subsequent tests as a are present, several methods are available for deter-
result of the destruction of the transfused RBCs, as mining the phenotype of only the patient’s RBCs:
was true in the case from Croucher and colleagues
that was described previously. This is true even 1. One method depends on separating the younger
though the DAT might remain weakly positive for RBCs (e.g., reticulocytes are presumed to be the
several months following a DHTR. In contrast, a rapid patient’s own RBCs).370-375
diminution in the strength of the DAT would not be 2. Another method utilizes flow cytometry.376-378 We
expected in AIHA except as a result of treatment, as believe that this method is much more reliable than
would be the case with the use of corticosteroids, the reticulocyte method.
immunosuppressive drugs, or splenectomy. 3. A recent approach is to use DNA typing.379-381

Antibody Specificity Additional Approaches


An important means of differentiating AIHA from a If the recipient’s DAT was known to be negative prior
DHTR relates to the specificity of the antibody(ies) to transfusion, or if the recipient’s RBCs are still avail-
present in the serum and in an RBC eluate. Some anti- able for the performance of the DAT and it is demon-
bodies that commonly cause DHTR reactions have not strated to be negative, this can be valuable information.
been found or have been reported only rarely as An abrupt change in the DAT from negative to positive
autoantibodies in AIHA. Examples are anti-K and is strong evidence that the patient has a DHTR rather
anti-Fya, which are encountered frequently in pub- than AIHA.
lished cases of DHTRs.356,362,363,368,369 Many autoanti- If it is possible to test the donor units of blood, this
bodies in warm antibody AIHA demonstrate procedure can be of significance if an antibody having
specificity within the Rh system, but even here, a dis- specificity that could be either an alloantibody or an
tinction between autoantibodies and alloantibodies autoantibody is detected (anti-E, for example). If, by
with Rh specificity is often possible. Whereas alloanti- chance, none of the donor units contains the E antigen,
bodies demonstrate truly specific reactions and give AIHA or an alloantibody-induced HTR caused by an
clearly negative reactions with cells lacking the appro- undetected antibody must be considered.
priate antigen, autoantibodies commonly demon- Thus, with careful serologic testing, it is possible to
strate “relative specificity.” That is, autoantibodies distinguish a DHTR from AIHA in almost all cases. If
that are described as having specificity within the Rh pretransfusion RBCs are not available (as, unfortu-
system react more strongly or to a higher titer against nately, is usually the case), and if the patient has an
RBCs bearing a particular Rh antigen, but they will alloantibody or mixture of alloantibodies with a broad
nevertheless react with RBCs lacking that antigen. range of reactivity, the distinction could be difficult.
Thus, a truly specific Rh antibody strongly suggests A final note of caution is that the acquired immune
that it is an alloantibody, whereas an antibody demon- hemolytic anemia might be neither a DHTR nor AIHA
strating “relative specificity” is characteristic of but instead could be drug-induced immune hemolytic
autoantibody. On the other hand, a mixture of an Rh anemia. In particular, cephalosporin drugs are used
alloantibody and an autoantibody reacting equally frequently in association with surgical procedures that
but less strongly with all normal RBCs will appear could require transfusion. In this setting, abrupt onset
similar to an autoantibody. A differentiation of these of a positive DAT and hemolysis could be misinter-
two possibilities would be difficult except by using preted as a DHTR or the sudden onset of AIHA. This
the patient’s pretransfusion RBCs for cell typing or for topic is discussed in detail in Chapter 8.
358 Immune Hemolytic Anemias

H. Bystander Immune Hemolysis

THE CONCEPT OF BYSTANDER A BRIEF HISTORY OF THE


IMMUNE CYTOLYSIS DEVELOPMENT OF THE CONCEPT OF
BYSTANDER IMMUNE CYTOLYSIS
Immune lysis of blood cells may occur in situations in
which accepted immunologic principles would not In 1965, Dameshek384 described instances in which an
lead one to expect hemolysis of the cells that are being exogenous factor (e.g., bacterial infection) caused an
destroyed. The following examples may be cited: immune response leading to an antigen-antibody
reaction on a cell, or which resulted in injury to the
1. When an RBC alloantibody causes destruction of cell via the development of immune complexes. He
transfused RBCs in a hemolytic transfusion reaction, referred to cells involved in such reactions as “inno-
one would not expect hemolysis of the patient’s cent bystander” cells. He emphasized that these re-
own RBCs. actions involved exogenous antigens leading to the
2. When ABO antibodies are produced by the donor’s development of endogenous disease and distin-
lymphocytes following hematopoietic cell trans- guished such reactions from true autoimmune disor-
plantation and cause hemolysis of the patient’s ders. Instead, he considered these reactions to be
RBCs, one would not expect that transfused group O temporary “allergic” reactions.
RBCs would be hemolyzed. (See the section on pas- Subsequently, a number of studies have investi-
senger lymphocyte syndrome in Chapter 12.) gated possible mechanisms for bystander immune
3. When a person who has been transfused or who cytolysis and, more recently, additional clinical set-
has been pregnant develops an alloantibody tings in which bystander immune cytolysis occurs
against the HPA-1a antigen (PlA1) on platelets, one have been recognized.382,383,385
would not expect that lysis of the patient’s own
HPA-1a-negative platelets would occur following
a subsequent transfusion. Complement-Mediated Lysis
4. When a patient is infected with malaria parasites of “Bystander Cells”
that enter RBCs and cause their disruption, one
would not expect immune lysis of RBCs that are In the 1960s and early 1970s, several groups of investi-
not infected. gators described a form of RBC cell lysis differentiated
5. When a patient develops an immune reaction to a from classical complement-mediated hemolysis by its
drug, a vaccination, or a micro-organism, one does occurrence in the absence of antibody on the cells.386-391
not expect the development of immune hemolytic That is, RBCs are lysed by complement even though
anemia. they are not involved in an antigen-antibody reaction.
6. When a patient receives a blood transfusion, one These investigators demonstrated that activated com-
does not expect the development of autoimmune plement components (C567) were generated by an
hemolytic anemia. immune reaction and that the activated components
could attach to normal RBC membranes, which are
In the foregoing examples, an alloimmune stimula- then lysed in the presence of late-reacting complement
tion results in an antibody that apparently causes lysis components. Thus, the hemolysis results from activa-
of the patient’s own RBCs. In many instances, the only tion of complement components at one site, which
RBC antibody detected is directed against RBC antigens leads to the lysis of normal cells at a distance. The term
not intrinsic to the patient’s RBCs. In other instances, most commonly used to describe this mechanism was
alloimmunization leads to the development of autoanti- “reactive hemolysis.” In these studies, the early-
bodies. We suggest that such examples of immune cell reacting components of complement (C1, C4, C2, and
lysis be grouped under the heading of bystander immune C3) were not required for the reaction. RBCs from
cytolysis. Accordingly, we have developed the following patients with PNH were exceptionally susceptible to
definition to encompass these occurrences. this mode of hemolysis,392,393 although hemolysis of
normal human RBCs also occurred.389
Subsequently, Salama and Mueller-Eckhardt193 and
Definition of Bystander Immune Cytolysis Ness and associates394 reported that autologous RBCs
Bystander immune cytolysis may be defined as were sensitized by complement during many DHTRs.
immune destruction of cells by antibody directed That is, DHTRs caused by antibodies to transfused
against an antigen that is not an unmodified intrinsic RBCs resulted in sensitization by complement compo-
component of the cell membrane.382,383 We also apply nents of autologous RBCs that did not contain the
the term bystander immune cytolysis when autoimmune antigen to which the causative antibody was directed.
hemolytic anemia occurs following exposure to Because C3 was detected on the patients’ RBCs, com-
alloantigens. plement sensitization of the autologous cells occurred
Unusual Aspects of Acquired Immune Hemolytic Anemias 359

by a mechanism that is different from classical reac- TABLE 9-18. CLINICAL SETTINGS IN WHICH
tive hemolysis. In neither study was an effort made to BYSTANDER IMMUNE CYTOLYSIS COULD OCCUR
determine whether autologous RBCs had a shortened
life span during the DHTRs. 1. The passenger lymphocyte syndrome, especially in association
Lack of Frequent Clinical Reports of Bystander with minor ABO-incompatible marrow transplants.
Immune Hemolysis. Although complement-medi- 2. The sickle cell hemolytic transfusion reaction syndrome.
ated bystander lysis was described in detail in experi- 3. Severe hemolytic transfusion reactions.
4. Posttransfusion purpura.
mental studies, instances of bystander immune 5. Immune hemolytic associated with infectious agents.
hemolysis in clinical medicine were not well docu- 6. Paroxysmal nocturnal hemoglobinuria.
mented. Yachnin and Ruthenberg392 suggested that 7. Drug-induced immune hemolytic anemia.
the hemolysis in patients with PNH—especially the 8. AIHA following transfusion.
9. AIHA following vaccination.
bouts of worsened hemolysis occurring during acute
infections—might occur by this mechanism. They also
suggested that such a mechanism might explain the
pathogenesis of other DAT-positive acquired immune
hemolytic anemias wherein complement but not anti- numerous other clinical examples were identified, and
body could be demonstrated on the patients’ RBCs. In a number of potential mechanisms were proposed.
addition, there were isolated reports, often incom-
pletely documented, of the destruction of autologous CLINICAL SETTINGS IN WHICH
RBCs during DHTRs.131,395-397 Because so few clinical BYSTANDER IMMUNE CYTOLYSIS MAY
reports suggesting bystander hemolysis were avail-
able, there was little interest in the phenomenon. OCCUR
The Passenger Lymphocyte Syndrome and Bystan-
der Immune Hemolysis. Our interest in bystander Bystander cytolysis could play an important role in im-
immune cytolysis was stimulated by observations of an mune lysis in a number of clinical entities (Table 9-18).
extraordinary group of patients who developed the The Passenger Lymphocyte Syndrome. The passen-
passenger lymphocyte syndrome following minor ger lymphocyte syndrome is reviewed briefly above
ABO-incompatible bone marrow transplantation using and in more detail in Chapter 12. In addition to the
matched-unrelated donors (see Chapter 12).398 patients previously cited who were reported by
Hemolysis in the passenger lymphocyte syndrome is Gajewski and colleagues,398 there have been a number
generally attributed to destruction of the patient’s of other reports of severe hemolysis in this setting,
incompatible RBCs by donor-derived anti-A and/or some of which strongly suggest the hemolysis of trans-
anti-B produced from “passenger” immunocompetent fused group O RBCs in addition to the patients’ own
donor lymphocytes that are infused with the marrow ABO-incompatible RBCs.399-405 These cases provide
product. In three such patients whom we described, strong evidence for bystander immune hemolysis, as
however, transfusion requirements for group O RBCs serologic studies have revealed only antibodies against
during the hemolytic episode clearly indicated that the the patient’s own group A or B RBCs, yet large volumes
amount of hemolysis that occurred was far in excess of transfused group O RBCs are hemolyzed as well.
of what could be explained on the basis of hemolysis of The sickle cell hemolytic transfusion reaction syn-
the patients’ RBCs alone.398 The total RBC volumes of drome is described in Chapter 14. A prominent feature
the patients at the onset of hemolysis were estimated as of the syndrome is the fact that the patient’s hemoglo-
being between 1592 mL and 2039 mL, whereas appro- bin level following transfusion can be significantly
ximately 4680 mL of washed group O RBCs (26 units) lower than was present prior to transfusion, thereby
had to be transfused in the subsequent 15 days to main- suggesting that, in addition to hemolysis of the trans-
tain a reasonable hemoglobin level. (These data are pre- fused RBCs, the patient’s own RBCs are involved in the
sented in detail in Chapter 12.) As the patients were not hemolytic reaction. The analysis is complicated by the
bleeding, these findings provided compelling evidence fact that a shortened RBC survival time is an invariable
that transfused group O RBCs were also hemolyzed. finding in SCD, and by the fact that reticulocytopenia
All of the patients developed strongly reactive anti-A or frequently occurs during the DHTR.406 Nevertheless, a
anti-B as is characteristic of the passenger lymphocyte frequently proposed mechanism for the profound drop
syndrome, and no other antibodies were detected in in hemoglobin that often occurs in this setting is an
their sera or in eluates from their RBCs. Because anti-A increased rate of destruction of the patient’s own RBCs
and anti-B were the only RBC antibodies present, yet (“hyperhemolysis”).382,406-410 Briefly stated, the
group O RBCs were also hemolyzed, we suggested that patient’s own RBCs are affected by the immune lysis of
this was an example of bystander immune hemolysis, allogeneic RBC. Although “hyperhemolysis” is most
in which the anti-A and anti-B indirectly caused the often described among patients with SCD who have
hemolysis of the group O RBCs, perhaps through a DHTRs, similar findings have been described among
mechanism such as reactive hemolysis. patients with thalassemia.411
These cases stimulated us to review and extend the Because “hyperhemolysis” resulting in a lower
concept of bystander immune cytolysis.382 In so doing, hemoglobin level following transfusion than that
360 Immune Hemolytic Anemias

prior to transfusion has been reported following fractions containing platelet material.413,414 At the time
DHTRs in patients with two forms of hereditary thrombocytopenia develops, an antibody against an
hemolytic anemia, one wonders whether a similar alloantigen on transfused platelets is present in the
occurrence might not sometimes follow DHTRs in patient’s serum. The alloantibody, usually anti-HPA-
patients with acquired hemolytic anemias.383 1a (anti-PlA1), develops as a result of prior transfusion
Severe hemolytic transfusion reactions in which the or pregnancy but appears to result in destruction of
patients’ own RBCs are also hemolyzed have been the patient’s HPA-1a-negative platelets. The mecha-
described infrequently. Polesky and Bove131 described nism of thrombocytopenia has remained enigmatic
a 72-year-old female who was hospitalized because of because the patient’s platelets after recovery are
severe anemia. On the patient’s fourth hospital day, an invariably nonreactive with alloantibody that is
RBC survival study using her own 51Cr-labeled RBCs present during the acute phase of PTP.415
was started because she had a positive DAT and a non- Several hypotheses have been proposed.416 Soluble
specific cold autoantibody with a wide thermal ampli- HPA-1a antigen on platelet membrane microparticles is
tude and had recently had a febrile transfusion present in blood products417 and might adsorb to the
reaction. Twelve days after the 51Cr-labeled RBC sur- patient’s platelets, providing target antigen.418,419 A
vival study was started, 300 mL of washed RBCs were second hypothesis is that immune complexes of soluble
transfused. No symptoms occurred during the transfu- HPA-1a antigen and anti-HPA-1a alloantibodies
sion, but about 1 hour later, the patient voided 145 mL mediate autologous platelet destruction.413,420 A third
port wine–colored urine. Hemoglobinuria and hemo- hypothesis is that an autoantibody forms in parallel
globinemia were documented, and her hematocrit, with the alloantibody, recognizing a conserved struc-
which had been stable at 18%–19% for 12 days, was tural determinant adjacent to the specific antigen poly-
11% on the day after transfusion. She developed anuria morphic site, and that this antibody then destroys
and increasing jaundice and expired 3 days later. autologous platelets.421
The RBC survival study demonstrated that 28% of AIHA associated with infectious agents might
the patient’s own RBCs were acutely hemolyzed. It is develop by a number of mechanisms that are reviewed
also probable that all of the transfused RBCs were in another segment of this chapter (see the section on
hemolyzed. The DHTR was attributed to an anti-Jka AIHA and infectious agents). These cases of AIHA are
that had been obscured by the presence of the autoan- further examples of bystander immune hemolysis.
tibody and was ultimately detected after the patient’s Paroxysmal nocturnal hemoglobinuria (PNH) is a
death. The authors suggested that the autoantibody disorder in which RBCs are particularly sensitive to
was the most probable cause of the hemolysis of the the action of complement. Bystander immune hemo-
patients’ own RBCs during the DHTR. They also cited lysis has been thought to contribute to at least some
two similar cases,395,396 but added that these cases had episodes of hemolysis.422
not been documented in detail. There are several abnormalities of PNH RBC mem-
Wiener397 described a patient whose hemoglobin branes that modulate complement, especially decay
dropped from 6 g/dL to 2.7 g/dL following transfusion accelerating factor (DAF, CD55) and the membrane
of 500 mL of blood that was subsequently determined inhibitor of reactive lysis (MIRL, CD59). It appears
to be incompatible. Signs and symptoms of hemolysis that absence of the CD59 antigen plays the most criti-
were present, and there was no evidence of blood loss. cal role in the complement sensitivity of PNH RBCs.
Accordingly, he concluded that transfusions of incom- Inherited deficiency of DAF is not associated with
patible blood could initiate some mechanism that clinical hemolysis, whereas a hereditary deficiency of
causes destruction of the patient’s own RBCs in addi- CD59 is associated with PNH. Restoration of CD59 in
tion to causing lysis of the donor’s RBCs. vitro corrects the complement sensitivity of RBCs
Greene and Khan412 described a 62-year-old female more completely than restoration of DAF, and PNH
whose pretransfusion testing demonstrated alloanti-c. can occur in the absence of DAF deficiency.422
During surgery, 5 units of c-negative RBCs were trans- Yachnin and Ruthenberg392 emphasized that their
fused, and 10 days later her hemoglobin plummeted experiments with PNH RBCs indicated that produc-
to 4.9 g/dL—a 9.6 g/dL drop. Serologic studies indi- tion of the RBC membrane damage by complement
cated a DHTR due to alloanti-s, -Fya, and -Jkb. The did not involve an antigen-antibody reaction specific
authors pointed out that the drop in hemoglobin was for the RBCs that were lysed. They suggested that the
greater than could be attributed to lysis of the trans- well-known propensity of patients with PNH to be
fused RBCs only and stated that it was evident that worsened by bouts of intercurrent infection might be
autologous RBCs were being hemolyzed as well. They related to immune interactions between antibody and
indicated that anti-Jkb might fix complement and con- the invading agent or its products that nonspecifically
cluded that complement activation occurring during (“indifferently”) activate the complement system.
the DHTR resulted in lysis of innocent bystander Rosse423 and Nakakuma and coworkers424,425 have
(autologous) RBCs. made similar suggestions.
Post-transfusion purpura (PTP) is characterized by Sirchia and colleagues426 demonstrated that when
sudden onset of severe thrombocytopenia approxi- PNH RBCs were incubated in vitro with leukocytes
mately 1 week following transfusion of blood or blood and a fresh, unacidified serum containing an anti-
Unusual Aspects of Acquired Immune Hemolytic Anemias 361

leukocyte antibody, hemolysis was observed, and the Lachmann387 and Lachmann and Thompson.388 As indi-
intensity of lysis varied according to the number of cated previously, they described a form of RBC lysis
leukocytes used. These researchers stated that their (which they termed reactive lysis) in which cells are lysed
findings supported a previous suggestion made by by complement even though they are not involved in an
Dacie427 that DHTRs that have been reported in some antigen-antibody reaction. They emphasized that this is
PNH patients428-435 might be caused by the interaction a mechanism by which complement activation at one
between transfused leukocytes and leukocyte antibo- site can lyse normal cells at a distance.388 Other in vitro
dies in the patients’ sera. Indeed, washed RBCs431,436 test systems demonstrated complement activation by
or white-cell poor RBCs426,437 have been recommended somewhat different mechanisms that also resulted in
for transfusion of patients with PNH to avoid DHTRs. sensitization of bystander cells.193,394,440
Other investigators, however, have suggested that this Experimental Data. Thompson and Rowe386 sus-
might not be necessary438 provided that group-specific pended antibody-sensitized sheep RBCs in agarose
blood products are transfused.439 gel and placed human serum as a source of comple-
Drug-induced immune hemolytic anemia is ment in wells cut in the gel. In addition to zones of
reviewed in depth in Chapter 8. A number of mecha- hemolysis surrounding the wells, they observed
nisms have been proposed by which the development linear zones of hemolysis beyond the initial hemolytic
of an immune response to a drug results in hemolysis. zones. They referred to the latter zones of hemolysis
As indicated previously, the resultant antibodies are as reactive hemolysis because they appeared as a
not directed against unmodified intrinsic RBC anti- result of the interaction of factors from different sera,
gens; they therefore satisfy our definition of bystander although it became apparent during their investiga-
immune hemolysis. tion that both factors could be obtained from the same
Autoimmune hemolytic anemia following trans- serum. They further determined that sensitization of
fusion is an example of an alloimmune stimulus the cells by antibody was not necessary for reactive
resulting in autoimmune hemolysis. Because the hemolysis and that sheep, rabbit, or human RBCs
patient’s own RBCs are hemolyzed as a result of an could be lysed in this system. Hemolysis also
alloimmune response, we include this as an example occurred even when EDTA was incorporated into the
of bystander immune hemolysis. This topic is plate, thus excluding the sequential activation of RBC
reviewed in another segment of this chapter (see the surface-bound complement components.
section on development of RBC autoantibodies and Reactive hemolysis was demonstrable following
AIHA following transfusion). “activation” of certain sera by agents known to acti-
Autoimmune Hemolytic Anemia Following vate complement, such as antibody-coated bacteria
Vaccination. This topic is reviewed in Chapter 3. and zymosan. Some activation procedures were espe-
cially effective in producing the lytic factors that
caused reactive hemolysis, and the authors suggested
POSSIBLE MECHANISMS OF that hemolysis was particularly likely to occur in cir-
BYSTANDER IMMUNE CYTOLYSIS cumstances in which serum inhibitors of complement
were not effective.
A number of possible mechanisms have been proposed Lachman and Thompson388 demonstrated that the
as the basis for bystander immune cytolysis (Table 9-19) complement factor generated by activation of serum
and are detailed in the following paragraphs.382 that was responsible for reactive hemolysis was C567
(a complex of complement factors 5, 6, and 7). This
Complement-Mediated Lysis complex is formed by the complexing of C7 with C56,
the latter being formed in the fluid phase after
of Unsensitized Cells immune activation. The investigators demonstrated
Reactive Hemolysis. Thompson and Rowe386 des- that C567 can attach to normal RBC membranes,
cribed a novel mechanism of cell lysis, and further which are then lysed in the presence of C8 and C9.
experimental data were published by Thompson and C567 can only be generated in a minority of sera, gen-
erally those from patients in the “acute phase” of
inflammation. They determined that much more C567
TABLE 9-19. POSSIBLE MECHANISMS is bound to cells when it is generated in solution
OF BYSTANDER IMMUNE CYTOLYSIS rather than at the complement fixation site (i.e., the
cell membrane in an antigen-antibody reaction), and
1. Complement mediated lysis of unsensitized cells. they suggested that this is due to the fact that there
a. Reactive hemolysis could be a much greater area of membrane available
b. Complement activation by mechanisms other than reactive
hemolysis. in the former case. They confirmed that the earlier-
2. Immune reactions against antigens adsorbed from plasma. reacting components of complement (C1, C4, C2, and
3. Adsorption of antigen-antibody complexes. C3) are not required for the reaction, although activa-
4. Modification of cellular antigens by drugs. tion of C3 by the classical immune hemolytic sequence
5. Development of RBC autoantibodies as a result of
alloimmunization.
utilizing C1, C4, and C2 can also be used to generate
C56. Although it is convenient to carry out reactive
362 Immune Hemolytic Anemias

hemolysis in immunodiffusion plates, the reaction complement components because of cross-reactivity of


works equally well in the test tube. alloantibodies with autologous RBCs, or as a result of
Götze and Muller-Eberhard389 also described fluid-phase complement activation. A later report by
mechanisms of cell lysis by complement that were Ness and coworkers394 also described sensitization of
entirely independent of antibody and that did not autologous RBCs in DHTRs. Because C3 was detected
require binding of the first four complement compo- on the patients’ RBCs, complement sensitization of the
nents to the target-cell surface. The actual attack of autologous cells occurred by a mechanism that is dif-
the target cell begins with the attachment of C5, C6, ferent from that of classical reactive hemolysis.
and C7. The binding reaction is catalyzed by C423, an Although Götze and Muller-Eberhard389 had failed
enzyme that might be formed in cell-free solution. to demonstrate C3 binding after activation of comple-
C423 might effect binding of C567 by acting from the ment at the surface of an adjacent cell, Salama and
fluid phase or from the surface of another cell to Mueller-Eckhardt440 presented evidence that C3 can
which it is bound specifically. The resulting cells are indeed be attached to bystander human RBCs if com-
susceptible to lysis by C8 and C9. Transfer of C567 plement is activated either through the classical
from the site of activation on one cell to the site of pathway or through the alternative pathway.
binding on another cell is distinct from any other The Role of Complement-Mediated Lysis of
known transfer phenomenon of complement compo- Unsensitized Cells as a Mechanism for Bystander
nents. The authors stated that this mechanism might Immune Cytolysis. PNH is a disorder in which such a
underline the phenomenon of reactive hemolysis mechanism would seem likely because of the markedly
reported by Thompson and Lachmann.387 They also increased sensitivity of the RBCs in this disorder to lysis
pointed out that PNH cells were exceptionally sus- by complement. Increased sensitivity to complement-
ceptible to this mode of hemolysis. mediated lysis has also been demonstrated in RBCs
Yachnin and Ruthenberg392 and Yachnin393 pre- from patients with sickle cell anemia,442 suggesting that
viously had described fluid phase activation of com- this mechanism might contribute to “hyperhemolysis”
plement resulting in the lysis of normal human RBCs in the sickle cell hemolytic transfusion reaction syn-
and of the RBCs from patients with PNH. They drome. Further, this mechanism might be operative in
pointed out that the demonstration that cells can patients who have immune hemolysis caused by ABO
suffer damage and destruction by the complement blood group antibodies, as these antibodies regularly
system without the mediation of specific anticell anti- cause activation of complement. This could occur
body suggests that autoimmune mechanisms need during the passenger lymphocyte syndrome following
not necessarily be invoked to explain the presence of ABO-incompatible hematopoietic cell transplantation,
complement components on cells. At the time of these or during HTRs caused by anti-A or anti-B.
earlier studies, the components of complement
causing the reaction were not as well defined, and Immune Reactions Against Antigens
these authors did not use the term reactive lysis, Adsorbed from Plasma by RBCs
although subsequent researchers have commented
that the underlying mechanisms seem similar.382,385 Adsorption of A and B Antigens. Because A and B
Other investigators also described various basic antigens can be demonstrated in plasma,443-445 one
aspects of the reaction mechanism of reactive hemoly- possible explanation for the hemolysis of transfused
sis.390,391 Goldman and colleagues390 reported that when group O RBCs in patients with the passenger lympho-
the average numbers of C567 sites per RBC generated in cyte syndrome caused by anti-A or anti-B is the
the presence of optimal C7 were plotted as a function of adsorption of the A or B antigen onto the transfused
C56 input, linear responses were obtained, indicating RBC. This could allow for hemolysis by the strongly
that a single competent C567 site on a RBC membrane reactive anti-A and anti-B produced by the passenger
is sufficient to prepare the cell for lysis by C8 and C9. lymphocytes in the donor stem cell product. This sce-
Gocke441 reported a similar system involving in vitro nario could be more likely in patients who are secre-
damage to platelets by the complement system, which tors than in patients who are nonsecretors because
is initiated by an antigen-antibody system in which the “nonsecretors” secrete very small amounts of A or B
antigen is not an integral part of the platelet. according to their ABO group.443,446
Complement Activation by Mechanisms Other There is a significant body of data indicating adsorp-
Than Reactive Hemolysis. Salama and Mueller- tion of A and B antigens onto group O RBCs following
Eckhardt193 performed detailed serologic studies on 26 transfusion, after ABO-incompatible hematopoietic cell
patients with DHTRs and found a positive DAT due to transplantation, or in congenital blood group chimeras.
C3d in all cases. The antiglobulin reactions were not of Renton and Hancock447 observed that when group O
the “mixed-field” type, which indicated that all of the RBCs are transfused to group A or group B recipients,
patients’ RBCs were coated with complement. The they can acquire small amounts of A or B substance.
authors concluded that complement is activated regu- The uptake of antigen is best demonstrated by using
larly in DHTRs and binds to both autologous RBCs and certain group O sera and to a lesser extent with group
the transfused cells. These researchers suggested that A and B sera. This phenomenon has been illustrated
the autologous RBCs could become sensitized with well by Crookston and Tilley443 in a patient who was
Unusual Aspects of Acquired Immune Hemolytic Anemias 363

FIGURE 9-9. In both pictures, the red cells are


from the same suspension of a patient’s blood
(16% A, 84% O). On the left, the cells are tested
with anti-A serum. On the right, the cells are tested
with anti-A,B (group O) serum.58 (From Szymanski
IO, Tilley CA, Crookston MC, Greenwalt TJ, Good
RA: Lewis substances in a human marrow-
transplantation chimaera. Lancet 1: 396, 1971.
Reprinted with permission from Elsevier.)

blood group chimera and had 16% group A1 RBCs and Adsorption of Other Antigens from Plasma. One
84% group O RBCs (Fig. 9-9). On the left, the RBCs are of the mechanisms proposed for lysis of HPLA-1a
tested with anti-A serum; on the right, they are tested negative platelets in patients with anti-HPA-1a is the
with anti-A,B (group O) serum. (This degree of aggluti- adsorption to the patient’s platelets of soluble HPA-1a
nation is much stronger than typically occurs after antigen, thereby providing target antigen. A similar
transfusion of group O RBCs to a group A patient.) mechanism could account for bystander immune
Other investigators also have reported that group O hemolysis associated with infectious agents. Further,
RBCs in blood group chimeras who are genetically in some instances, drug administration might cause
group A are agglutinated by group O sera.448-450 adsorption of drug antigens to the RBC membrane,
Although routine ABO blood grouping following with lysis mediated by antidrug antibody.
ABO-incompatible marrow transplantation generally
indicates conversion to donor type, we have observed Adsorption of Antigen-Antibody
weakly positive agglutination reactions a year or
longer after marrow transplantation using anti-A,B
Complexes
sera when testing RBCs from patients who had been Another mechanism that could account for the hemo-
group A prior to transplantation with a group O lysis of group O RBCs in minor ABO-mismatched trans-
marrow.382 Also, Crookston and Tilley443 reported on plant patients is the adsorption of antigen-antibody
three patients who were tested about two years after complexes. It is possible that the anti-A or anti-B pro-
receiving a bone marrow transplant from a group O duced by the passenger lymphocytes reacts with A or B
donor. In two group A1, Le(a-b+) recipients, the grafted antigen in the plasma and that these antigen-antibody
group O RBCs reacted strongly with anti-A,B sera. In a complexes are adsorbed by the transfused group O
group A1, Le(a+b-) recipient, the grafted group O RBCs RBCs. Further, destruction of antigen-negative cells as a
reacted weakly with anti-A,B sera. Further, Maeda and result of adsorption of antigen-antibody complexes has
coworkers451 found positive DAT reactions and could been proposed as a possible mechanism responsible in
elute anti-B from the RBCs of a patient who had been some instances of drug-induced immune hemolysis,
group B and had received a transplant from a group A and in the destruction of HPA-1a negative platelets in
person. They suggested that the most likely explana- post-transfusion purpura. Also, in infectious diseases,
tion was that anti-B was produced by the donor antibody-antigen complexes specifically related to the
marrow and reacted with B antigen adsorbed from the infectious agent might coat the RBC, which then act as
patient’s plasma onto the group B RBCs produced by an “innocent bystander.” Similarly, parasite antigen-
the transplanted stem cells. antibody immune complexes likely contribute to the
As reviewed in Chapter 12, Arndt and associates452 pathogenesis of the anemia in falciparum malaria. In
used flow cytometry to demonstrate that the recip- some autoimmune disorders, particularly SLE, immune
ient’s RBCs following hematopoietic cell transplanta- complexes frequently are detected in the patient’s
tion of group A or B patients with group O stem cells plasma and could contribute to the frequent finding of
were uniformly coated with A or B substance. These hemolysis in these patients. Further, several groups of
findings are consistent with the concept of adsorption investigators have indicated that immune complexes
of group A and B substance rather than a mixture of could play an important role in removing transfused
group A and O RBCs. platelets from the circulation.453-456
364 Immune Hemolytic Anemias

Modification of Cellular Antigens by Drugs 2. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Warm-
antibody syndromes VI: Coombs-negative (DAT-negative)
The possible mechanisms by which the administration haemolytic anaemia; positive direct antiglobulin tests in
of drugs might cause immune hemolysis are reviewed normal subjects and in hospital patients; polyagglutinability
and haemolytic anaemia. In The Haemolytic Anaemias, 3rd
in Chapter 8. Prominent among the proposed mecha- ed. Vol. 3, The Autoimmune Haemolytic Anaemias. New
nisms is the development of antibodies that are York: Churchill Livingstone, 1992:183–209.
directed against cellular antigens modified by the drug. 3. Worlledge SM, Blajchman MA: The autoimmune haemolytic
anaemias. Br J Haematol 1972;23(S):61–69.
4. Chaplin H, Jr: Clinical usefulness of specific antiglobulin
reagents in autoimmune hemolytic anemias. Progr Hematol
Development of RBC Autoantibodies 1973;8:25–49.
as a Result of Alloimmunization 5. Boccardi V, Girelli G, Perricone R, Ciccone F, Romoli P, Isacchi
G: Coombs-negative autoimmune hemolytic anemia: Report
AIHA following transfusion is an example of alloim- of 11 cases. Haematologica 1978;63:301–310.
munization leading to an autoimmune reaction457 6. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias,
1st ed. New York: Churchill Livingstone, 1980.
(and see Section B of this chapter). The mechanism by 7. Gilliland BC, Leddy JP, Vaughan JH: The detection of cell-
which this occurs is uncertain, but other possible bound antibody on complement-coated human red cells.
examples include the development of AIHA associ- J Clin Invest 1970;49:898–906.
ated with infection and vaccination. These topics are 8. Gilliland BC, Baxter E, Evans RS: Red-cell antibodies in
reviewed in Chapter 3. acquired hemolytic anemia with negative antiglobulin serum
tests. N Engl J Med 1971;285:252–256.
9. Gilliland BC: Coombs-negative immune hemolytic anemia.
Seminars Hematol 1976;13:267–275.
Lack of Specificity of Alloantibodies 10. Mollison PL, Hughes-Jones NC: Clearance of Rh-positive red
(“Cross-Reacting” Alloantibodies) cells by low concentrations of Rh antibody. Immunology
1967;12:63–73.
There is evidence that alloantibodies are not as exquis- 11. Constatoulakis M, Costea N, Schwartz RS, Dameshek W:
itely specific as appears to be the case in routine sero- Quantitative studies of the effect of red blood cell sensitization
in vitro hemolysis. J Clin Invest 1963;42:1790–1801.
logic reactions.385 The work of Marks and colleagues,458 12. Jandl JH, Kaplan ME: The destruction of red cells by anti-
Ouwehand and coworkers,459 Thompson and associ- bodies in man. III: Quantitative factors influencing the
ates,460 and Barker and Elson461 at the molecular level patterns of hemolysis in vivo. J Clin Invest 1960;39:1145.
suggests that perhaps all alloantibodies are capable of 13. Garratty G, Nance SJ: Correlation between in vivo hemolysis
and the amount of red cell-bound IgG measured by flow
reacting, under some circumstances, with RBCs lacking cytometry. Transfusion 1990;30:617–621.
the putative antigens. 14. Garratty G: Effect of cell-bound proteins on the in vivo sur-
Salama and Mueller-Eckhardt193 and Ness and asso- vival of circulating blood cells. Gerontology 1991;37:68–94.
ciates394,462 have suggested that one possible mecha- 15. Garratty G: Factors affecting the pathogenicity of red cell
nism involved in the sensitization of autologous RBCs auto- and alloantibodies. In Nance SJ (ed): Immune
Destruction of Red Blood Cells. Arlington, VA: American
with complement during a HTR could be the presence Association of Blood Banks, 1989:109–169.
of “cross-reacting” alloantibodies. In two patients with 16. Garratty G: The significance of IgG on the red cell surface.
DHTRs, Salama and Mueller-Eckhardt193 were able to Transfus Med Rev 1987;1:47–57.
adsorb alloanti-E from the serum with E-negative RBCs. 17. Merry AH, Thomson EE, Rawlinson VI, Stratton F: A quanti-
tative antiglobulin test for IgG for use in blood transfusion
This lack of definitive specificity could also be the expla- serology. Clin Lab Haematol 1982;4:393–402.
nation for “mimicking antibodies”154–156 (i.e., alloanti- 18. Jeje MO, Blajchman MA, Steeves K, Horsewood P, Kelton JG:
bodies that react with antigen-negative RBCs).385 Quantitation of red cell-associated IgG using an immunora-
Morrison and Mollison464 suggested that in cases of diometric assay. Transfusion 1984;24:473–476.
PTP, a weaker cross-reacting antibody (which they 19. Masouredis SP, Branks MJ, Victoria EJ: Antiidiotypic IgG
crossreactive with Rh alloantibodies in red cell autoimmunity.
referred to as an autoantibody) is formed, in addition to Blood 1987;70:710–715.
the allo-HPA-1a, and that it is this weaker antibody 20. Masouredis SP, Branks MJ, Garratty G, Victoria EJ:
that reacts with the HPA-1a-negative platelets. Immunospecific red cell binding of iodine 125-labeled
The lack of definitive specificity of alloantibodies immunoglobulin G erythrocyte autoantibodies. J Lab Clin
Med 1987;110:308–317.
might explain the presence of complement compo- 21. Dupuy ME, Elliot M, Masouredis SP: Relationship between
nents on autologous RBCs more than 100 days after red cell bound antibody and agglutination in the antiglobulin
the reaction, as has been reported by Salama and reaction. Vox Sang 1964;9:40–44.
Mueller-Eckhardt193 and by Ness and associates.394,462 22. Hughes-Jones NC, Polley MJ, Telford R, et al: Optimal condi-
The presence of cross-reacting alloantibodies could be tions for detecting blood group antibodies by the antiglobulin
test. Vox Sang 1964;9:385–395.
the explanation for some instances of bystander 23. Romano EL, Hughes-Jones NC, Mollison PL: Direct antiglob-
immune cytolysis. ulin reaction in ABO-haemolytic disease of the newborn. Br
Med J 1973;1:524–526.
24. Schmitz N, Djibey I, Kretschmer V, Mahn I, Mueller-Eckhardt
C: Assessment of red cell autoantibodies in autoimmune
R E F E R E N C E S hemolytic anemia of warm type by a radioactive anti-IgG test.
Vox Sang 1981;41:224–230.
1. Evans RS, Weiser RS: The serology of autoimmune hemolytic 25. Yam P, Petz LD, Spath P: Detection of IgG sensitization of red
disesase: Observations on forty-one patients. Arch Intern Med cells with 125I staphylococcal protein A. Am J Hematol 1982;
1957;100:371–399. 12:337–346.
Unusual Aspects of Acquired Immune Hemolytic Anemias 365

26. Salama A, Mueller-Eckhardt C, Bhakdi S: A two-stage 47. Petz LD, Yam P, Wilkinson L, Garratty G, Lubin B, Mentzer W:
immunoradiometric assay with 125I-staphylococcal protein A Increased IgG molecules bound to the surface of red blood
for the detection of antibodies and complement on human cells of patients with sickle cell anemia. Blood 1984;
blood cells. Vox Sang 1985;48:239–245. 64:301–304.
27. Nilsson EK, Loof L, Nilsson UR, Nilsson B: Development of 48. Kay MM: Aging of cell membrane molecules leads to appear-
an immunoassay for the detection of minute amounts of IgG- ance of an aging antigen and removal of senescent cells.
coated erythrocytes in whole blood and its application for the Gerontology 1985;31:215–235.
assessment of Fc-mediated clearance of anti-D-coated ery- 49. Green GA, Rehn MM, Kalra VK: Cell-bound autologous
throcytes in vivo. Vox Sang 1989;57:188–192. immunoglobulin in erythrocyte subpopulations from patients
28. Garratty G, Postoway N, Nance S, et al: The detection of IgG with sickle cell disease. Blood 1985;65:1127–1133.
on the red cells of “Coombs-negative” autoimmune hemolytic 50. Green GA, Kalra VK: Sickling-induced binding of immuno-
anemias [abstract]. Transfusion 1982;22: 430. globulin to sickle erythrocytes. Blood 1988;71:636–639.
29. Bodensteiner D, Brown P, Skikne B, Plapp F: The enzyme- 51. Hebbel RP, Miller WJ: Phagocytosis of sickle erythrocytes:
linked immunosorbent assay: Accurate detection of red blood Immunologic and oxidative determinants of hemolytic
cell antibodies in autoimmune hemolytic anemia. Am J Clin anemia. Blood 1984;64:733–741.
Pathol 1983;79:182–185. 52. Solanki DL: Erythrophagocytosis in vivo in sickle cell anemia.
30. Hasselbalch H, Berild D, Hansen OP: Red-cell sensitization in Am J Hematol 1985;20:353–357.
myelofibrosis. Scand J Haematol 1984;32:179–182. 53. Galili U, Korkesh A, Kahane I, Rachmilewitz EA: Demon-
31. Leach M: A direct enzyme-linked antiglobulin test for detec- stration of a natural antigalactosyl IgG antibody on thal-
tion of red cell autoantibodies in auto-immune haemolytic assemic red blood cells. Blood 1983;61:1258–1264.
anaemia. Med Lab Sci 1984;41:232–237. 54. Toy PT, Chin CA, Reid ME, Burns MA: Factors associated with
32. Hansen OP, Hansen TM, Jans H, Hippe E: Red blood cell positive direct antiglobulin tests in pretransfusion patients: A
membrane-bound IgG: Demonstration of antibodies in case-control study. Vox Sang 1985;49:215–220.
patients with autoimmune haemolytic anaemia and immune 55. Heddle NM, Kelton JG, Turchyn KL, Ali MA: Hypergam-
complexes in patients with rheumatic diseases. Clin Lab maglobulinemia can be associated with a positive direct
Haematol 1984;6:341–349. antiglobulin test, a nonreactive eluate, and no evidence of
33. Postoway N, Nance SJ, Garratty G: Variables affecting the hemolysis. Transfusion 1988;28:29–33.
enzyme-linked antiglobulin test when detecting and quanti- 56. Garratty G, Arndt P: Is there a correlation with RBC-bound
tating IgG red cell antibodies. Med Lab Sci 1985;42:11–19. IgG detected by sensitive assays, such as the enzyme-linked
34. Sokol RJ, Hewitt S, Booker DJ, Stamps R: Enzyme linked antiglobulin test (ELAT), and serum levels of IgG? [abstract]
direct antiglobulin tests in patients with autoimmune haemol- Transfusion 1987;27:545.
ysis. J Clin Pathol 1985;38:912–914. 57. Engelfriet CP, von dem Borne AEG, Beckers D, et al: Immune
35. Sokol RJ, Hewitt S, Booker DJ, Stamps RL: Small quantities of destruction of red cells. In Bell CA (ed): A Seminar on
erythrocyte bound immunoglobulins and autoimmune Immune-Mediated Cell Destruction. Washington, DC:
haemolysis. J Clin Pathol 1987;40:254–257. American Association of Blood Banks, 1981:93–130.
36. Howard PR, MacPherson B: An antiglobulin consumption 58. Zupanska B, Brojer E, Maslanka K, Hallberg T: A comparison
ELAT technique for the detection of red cell antibodies. J Med between Fc receptors for IgG1 and IgG3 on human monocytes
Technol 1987;4:218–222. and lymphocytes using anti-Rh antibodies. Vox Sang
37. Gutgsell NS, Issitt PD, Tomasulo PA, et al: Use of the direct 1985;49:67–76.
enzyme-linked antiglobulin test (ELAT) in patients with unex- 59. Garratty G, Arndt P, Nance S, et al: Low affinity autoanti-
plained anemia [abstract]. Transfusion 1988;28:36S. bodies—A cause of false negative direct antiglobulin tests
38. Tomasulo PA, Issitt PD, Gutgsell NS, et al: Predictive value of [abstract]. Book of Abstracts of the ISBT/AABB Joint
the direct enzyme-linked antiglobulin test (ELAT) in patients Congress, 1990:87.
with unexplained anemia [abstract]. London, Proceedings of 60. Garratty G: Low affinity autoantibodies—A cause of false
the 20th Congress of the International Society of Blood negative direct antiglobulin tests. Transfus Today 1991;
Transfusion, 1988:121. 12:3–4.
39. Kiruba R, Han P: Quantitation of red cell-bound immunoglo- 61. Sokol RJ, Booker DJ, Stamps R, et al: Direct Coombs test-
bulin and complement using enzyme-linked antiglobulin negative autoimmune hemolytic anemia and low-affinity IgG
consumption assay. Transfusion 1988;28:519–524. class antibodies. Immunohem 1997;13:115–118.
40. Garratty G, Postoway N, Nance S, Arndt P: Detection of IgG on 62. Lai M, Rumi C, D’Onofrio G, et al: Clinically significant
red cells of patients with suspected direct antiglobulin test autoimmune hemolytic anemia with a negative direct
negative autoimmune hemolytic anemia (AIHA) [abstract]. antiglobulin test by routine tube test and positive by column
Book of Abstracts of the ISBT/AABB Joint Congress, Los agglutination method. Immumnohematology 2002;18:109–113.
Angeles, 1990: 87. 63. Johnson ST, Bandouveres S: DAT negative autoimmune
41. Garratty G: Autoimmune hemolytic anemia. In Garratty G hemolytic anemia due to low affinity IgG warm-reactive anti-
(ed): Immunolobiology of Transfusion Medicine. New York: Pr in an infant [abstract]. Transfusion 2002;42:20S.
Dekker, 1994:493–521. 64. Schwartz RS, Costea N: Autoimmune hemolytic anemia:
42. Garratty G, Arndt P: Applications of flow cytofluorometry to Clinical correlations and biological implications. Semin
transfusion science. Transfusion 1995;35:157–178. Hematol 1966;3:2–26.
43. Wang Z, Shi J, Zhou Y, Ruan C: Detection of red blood cell- 65. Fossati-Jimack L, Reininger L, Chicheportiche Y, et al: High
bound immunoglobulin G by flow cytometry and its applica- pathogenic potential of low-affinity autoantibodies in experi-
tion in the diagnosis of autoimmune hemolytic anemia. Int J mental autoimmune hemolytic anemia. J Exp Med
Hematol 2001;73:188–193. 1999;190:1689–1696.
44. Rand BP, Olson JD, Garratty G, Petz LD: Coombs’ negative 66. Sturgeon P, Smith LE, Chun HMT, Hurvitz CH, Garratty G,
immune hemolytic anemia with anti-E occurring in the red Goldfinger D: Autoimmune hemolytic anemia associated
blood cell eluate of an E-negative patient. Transfusion exclusively with IgA of Rh specificity. Transfusion 1979;
1978;18:174–180. 19:324–328.
45. Ganly PS, Laffan MA, Owen I, Hows JM: Auto-anti-Jka in 67. Arndt P, Garratty G: Use of flow cytometry for detection of
Evans’ syndrome with negative direct antiglobulin test. Br J IgM on RBCs showing spontaneous agglutination [abstract].
Haematol 1988;69:537–539. Transfusion 1994;34:68S.
46. Yam P, Wilkinson L, Petz LD, Garratty G: Studies on 68. Garratty G, Arndt P, Domen R, et al: Severe autoimmune
hemolytic anemia in pregnancy with evidence for autoimmu- hemolytic anemia associated with IgM warm autoantibodies
nization in a patient with a negative direct antiglobulin directed against determinants on or associated with gly-
(Coombs’) test. Am J Hematol 1980;8:23–29. cophorin A. Vox Sang 1997;72:124–130.
366 Immune Hemolytic Anemias

69. Garratty G, Arndt P, Leger R: Serological findings in autoim- 91. Garratty G, Brunt D, Greenfield B, et al: An auto anti-Ena
mune hemolytic anemia associated with IgM warm autoanti- mimicking an allo anti-Ena associated with pure red cell
bodies [abstract]. Blood 2001;98:61a. aplasia [abstract]. Transfusion 1983;23:408.
70. Talano J-AM, Johnson ST, Friedman KD, et al: Serologic char- 92. Szymanski IO, Huff SR, Selbovitz LG, Sherwood GK:
acteristics of three cses of IgA mediated autoimmune Erythrocyte sensitization with monomeric IgM in a patient
hemolytic anemia (AIHA) confirmed by flow cytometry with hemolytic anemia. Am J Hematol 1984;17:71–77.
[abstract]. Blood 2002;100:283a. 93. Schreiber AD, Frank MM: Role of antibody and complement
71. Wager O, Haltia K, Rasanen JA, Vuopio P: Five cases of posi- in the immune clearance and destruction of erythrocytes. I: In
tive antiglobulin test involving IgA warm type autoantibody. vivo effects of IgG and IgM complement-fixing sites. J Clin
Ann Clin Res 1971;3:76–85. Invest 1972;51:575–582.
72. Stratton F, Rawlinson VI, Chapman SA, Pengelly CD, 94. Schreiber AD, Frank MM: Role of antibody and complement in
Jennings RC: Acquired hemolytic anemia associated with IgA the immune clearance and destruction of erythrocytes. II:
anti-e. Transfusion 1972;12:157–161. Molecular nature of IgG and IgM complement-fixing sites and
73. Suzuki S, Amano T, Mitsunaga M, Yagyu F, Ofuji T: effects of their interaction with serum. J Clin Invest 1972;
Autoimmune hemolytic anemia associated with IgA autoanti- 51:583–589.
body. Clin Immunol Immunopathol 1981;21:247–256. 95. Atkinson JP, Frank MM: Studies on the in vivo effects of anti-
74. Wolf CF, Wolf DJ, Peterson P, Brandstetter RD, Hansen DE: body. Interaction of IgM antibody and complement in the
Autoimmune hemolytic anemia with predominance of IgA immune clearance and destruction of erythrocytes in man.
autoantibody. Transfusion 1982;22:238–240. J Clin Invest 1974;54:339–348.
75. Clark DA, Dessypris EN, Jenkins DE, Jr, Krantz SB: Acquired 96. Cutbush M, Mollison PL: Relation between characteristics of
immune hemolytic anemia associated with IgA erythrocyte blood-group antibodies in vitro and associated patterns of red
coating: Investigation of hemolytic mechanisms. Blood cell destruction in vivo. Br J Haematol 1958;4:115–137.
1984;64:1000–1005. 97. Holburn AM, Frame M, Hughes-Jones NC, Mollison PL: Some
76. Kowal-Vern A, Jacobson P, Okuno T, Blank J: Negative direct biological effects of IgM anti-Rh (D). Immunology 1971;
antiglobulin test in autoimmune hemolytic anemia. Am J 20:681–691.
Pediatr Hematol Oncol 1986;8:349–351. 98. Mollison PL: Blood Transfusion in Clinical Medicine, 8th ed.
77. Reusser P, Osterwalder B, Burri H, Speck B: Autoimmune Oxford: Blackwell Scientific Publications, 1987.
hemolytic anemia associated with IgA—Diagnostic and ther- 99. Mollison PL: Survival curves of incompatible red cells: An
apeutic aspects in a case with long-term follow-up. Acta analytical review [Review]: Transfusion 1986;26:43–50.
Haematol 1987;77:53–56. 100. Salama A, Mueller-Eckhardt C: Autoimmune haemolytic
78. Salama A, Bhakdi S, Mueller-Eckhardt C: Evidence suggesting anaemia in childhood associated with non-complement
the occurrence of C3-independent intravascular immune hemol- binding IgM autoantibodies. Br J Haematol 1987;65:67–71.
ysis. Reactive hemolysis in vivo. Transfusion 1987;27:49–53. 101. Fabijanska-Mitek J, Namirska-Krzton H, Seyfried H: The
79. Gottsche B, Salama A, Mueller-Eckhardt C: Autoimmune value of gel test and ELAT in autoimmune haemolytic
hemolytic anemia associated with an IgA autoanti-Gerbich. anaemia. Clin Lab Haematol 1995;17:311–316.
Vox Sang 1990;58:211–214. 102. Kroll H, Salama A, Berghofer H, et al: The direct antiglobulin
80. Girelli G, Perrone MP, Adorno G, et al: A second example of test: Clinical significance and prospective comparison of
hemolysis due to IgA autoantibody with anti-e specificity. column agglutination technology, gel test and tube test in 163
Haematologica 1990;75:182–183. patients [abstract]. Vox Sanguinis 1994;67(S2):65.
81. Sokol RJ, Booker DJ, Stamps R, Booth JR, Hook V: IgA red cell 103. Sajur J, Fournier Y, Sheridan B: Comparison of gel and tube
autoantibodies and autoimmune hemolysis. Transfusion techniques for direct antiglobulin tests [abstract]. Transfusion
1997;37:175–181. 1997;37:27S.
82. Janvier D, Sellami F, Missud F, et al: Severe autoimmune 104. Petz LD, Branch DR: Serological tests for the diagnosis of
hemolytic anemia caused by a warm IgA autoantibody immune hemolytic anemias. In McMillan R (ed): Methods in
directed against the third loop of band 3 (RBC anion-exchange Haematology: Immune Cytopenias. New York: Churchill
protein 1). Transfusion 2002;42:1547–1552. Livingstone, 1983:9–48.
83. Bardill B, Mengis C, Tschopp M, Wuillemin WA: Severe IgA- 105. Owen I, Hows J: Evaluation of the manual hexadimethrine
mediated auto-immune haemolytic anaemia in a 48-yr-old bromide (Polybrene) technique in the investigation of autoim-
woman. Eur J Haematol 2003;70:60–63. mune hemolytic anemia. Transfusion 1990;30:814–818.
84. Fanger MW, Goldstine SN, Shen L: Cytofluorographic analy- 106. Rubino M, Kavitsky DM, Nance S: Serologic testing in
sis of receptors for IgA on human polymorphonuclear cells autoimmune hemolytic anemia (AIHA) with negative direct
and monocytes and the correlation of receptor expression antiglobulin test (DAT) [abstract]. Transfusion 2002;42:104S.
with phagocytosis. Mol Immunol 1983;20:1019–1027. 107. Garratty G, Petz LD: Acquired hemolytic anemia associated
85. Shen L, Maliszewski CR, Rigby WF, Fanger MW: IgA- with negative direct antiglobulin tests but enzyme reactive
mediated effector function of HL-60 cells following treatment autoantibodies in the serum [abstract]. Book of Abstracts of
with calcitriol. Mol Immunol 1986;23:611–618. the 24th Annual Meeting of the American Association of
86. Yeaman GR, Kerr MA: Opsonization of yeast by human serum Blood Banks, 1971:83.
IgA anti-mannan antibodies and phagocytosis by human poly- 108. van’t Veer MB, van Wieringen PM, van L, I, Overbeeke MA,
morphonuclear leucocytes. Clin Exp Immunol 1987;68:200–208. dem Borne AE, Engelfriet CP: A negative direct antiglobulin
87. Shen L, Fanger MW: Secretory IgA antibodies synergize with test with strong IgG red cell autoantibodies present in the
IgG in promoting ADCC by human polymorphonuclear serum of a patient with autoimmune haemolytic anaemia. Br
cells, monocytes, and lymphocytes. Cell Immunol 1981;59: J Haematol 1981;49:383–386.
75–81. 109. Issitt PD, Gruppo RA, Wilkinson SL, Issitt CH: Atypical pres-
88. Maliszewski CR, March CJ, Schoenborn MA, Gimpel S, Shen L: entation of acute phase, antibody-induced haemolytic
Expression cloning of a human Fc receptor for IgA. J Exp Med anaemia in an infant. Br J Haematol 1982;52:537–543.
1990;172:1665–1672. 110. Parker AC, Habeshaw J, Cleland JF: The demonstration of a
89. Kay NE, Douglas SD, Mond JJ, Flier JS, Kochwa S, Rosenfield “plasmatic factor” in a case of Coombs’ negative haemolytic
RE: Hemolytic anemia with serum and erythrocyte-bound anaemia. Scand J Haematol 1972;9:318–321.
low-molecular-weight IgM. Clin Immunol Immunopathol 111. van der Meulen FW, van der Hart M, Fleer A, Von dem Borne
1975;4:216–225. AE, Engelfriet CP, Van Loghem JJ: The role of adherence to
90. Schanfield MS, Pisciotta A, Libnock J: Seven cases of human mononuclear phagocytes in the destruction of red
hemolytic anemia associated with warm IgM autoantibodies cells sensitized with non-complement binding IgG antibodies.
[abstract]. Transfusion 1978;18:623. Br J Haematol 1978;38:541–549.
Unusual Aspects of Acquired Immune Hemolytic Anemias 367

112. Galili U, Manny N, Izak G: EA rosette formation: A simple 135. Beard ME, Pemberton J, Blagdon J, Jenkins WF: Rh immu-
means to increase sensitivity of the antiglobulin test in nization following incompatible blood transfusion and a pos-
patients with anti red cell antibodies. Br J Haematol 1981; sible long-term complication of anti-D immunoglobulin
47:227–233. therapy. J Med Genet 1971;8:317–320.
113. Nance S, Garratty G: Correlations between an in vitro mono- 136. Lalezari P, Talleyrand NP, Wenz B, Schoenfeld ME, Tippett P:
cyte monolayer assay and autoimmune hemolytic anemia Development of direct antiglobulin reaction accompanying
(AIHA) [abstract]. Transfusion 1982;22:410. alloimmunization in a patient with Rhd (D, category III) phe-
114. Gallagher MT, Branch DR, Mison A, Petz LD: Evaluation of notype. Vox Sang 1975;28:19–24.
reticuloendothelial function in autoimmune hemolytic 137. Worlledge SM: The interpretation of a positive direct antiglob-
anemia using an in vitro assay of monocyte-macrophage ulin test. Brit J Haemat 1978;39:157–162.
interaction with erythrocytes. Exp Hematol 1983;11:82–89. 138. Chaplin H Jr, Zarkowsky HS: Combined sickle cell disease
115. Brojer E, Zupanska B, Michalewska B: Adherence to human and autoimmune hemolytic anemia. Arch Intern Med
monocytes of red cells from autoimmune haemolytic anaemia 1981;141:1091–1093.
and red cells sensitized with alloantibodies. Haematologia 139. Sosler SD, Perkins JT, Saporito C, Unger P, Koshy M: Severe
(Budapest) 1982;15:135–145. autoimmune hemolytic anemia induced by transfusion in two
116. Zupanska B, Brojer E, Thomson EE, Merry AH, Seyfried H: alloimmunized patients with sickle cell disease [abstract].
Monocyte-erythrocyte interaction in autoimmune haemolytic Transfusion 1989;29:49S.
anaemia in relation to the number of erythrocyte-bound IgG 140. Argiolu F, Diana G, Arnone M, Batzella MG, Piras P, Cao A:
molecules and subclass specificity of autoantibodies. Vox High-dose intravenous immunoglobulin in the management
Sang 1987;52:212–218. of autoimmune hemolytic anemia complicating thalassemia
117. Herron R, Clark M, Young D, Smith DS: Correlation of major. Acta Haematol 1990;83:65–68.
mononuclear phagocyte assay results and in vivo haemolytic 141. Chan D, Poole GD, Binney M, Hamon MD, Copplestone JA,
rate in subjects with a positive direct antiglobulin test. Clin Prentice AG: Severe intravascular hemolysis due to autoanti-
Lab Haematol 1986;8:199–207. bodies stimulated by blood transfusion. Immunohematology
118. Garratty G: Basic mechanisms of in vivo cell destruction. In 1996;12:80–83.
Bell CA (ed): A Seminar on Immune-Mediated Cell 142. Zumberg MS, Procter JL, Lottenberg R, Kitchens CS, Klein
Destruction. Bethesda, Md: American Association of Blood HG: Autoantibody formation in the alloimmunized red blood
Banks, 1981:1–28. cell recipient: Clinical and laboratory implications. Arch
119. Gilsanz F, De La SJ, Molto L, Alvarez-Mon M: Hemolytic Intern Med 2001;161:285–290.
anemia in chronic large granular lymphocytic leukemia of 143. Szymanski IO, Smith VC: Red Blood Cell (RBC) autoimmunity
natural killer cells: Cytotoxicity of natural killer cells against in alloimmunized patients: Clinical observations [abstract].
autologous red cells is associated with hemolysis. Transfusion Blood 2001;98:111b.
1996;36:463–466. 144. Mohn JF, Lambert RM, Bowman HS, et al: Experimental pro-
120. Rous P, Robertson OH: Free antigen and antibody circulat- duction in man of autoantibodies with Rh specificity. Ann NY
ing together in large amounts (hemagglutinin and agglu- Acad Sci 1965;124:477.
tinogen in the blood of transfused rabbits). J Exp Med 145. Mohn JF, Bowman HS, Lambert RM, Brason FW: The forma-
1918;27:509–517. tion of Rh specific autoantibodies in experimental isoimmune
121. Ovary Z, Spiegelman J: The production of “cold autoagglu- hemolytic anemia in man. Tenth Congress of the International
tinins” in the rabbit as a consequence of immunization with Society for Blood Transfusion, Stockholm, 1964.
isologous erythrocytes. Ann N Y Acad Sci 1965;124:147–153. 146. Wodzinski MA, Collin RC, Booker DJ, Stamps R, Bellamy JD,
122. Zmijewski CM: The production of erythrocyte autoantibodies Sokol RJ: Delayed hemollytic transfusion reaction and par-
in chimpanzees. J Exp Med 1965;121:657. oxysmal cold hemoglobinuria: An unusual association.
123. Liu CK, Evans RS: Production of positive antiglobulin serum Immunohematology, 1997;13:54.
test in rabbits by intraperitoneal injection of homologous 147. Castellino SM, Combs MR, Zimmerman SA, Issitt PD, Ware
blood. Proc Soc Exp Biol Med 1952;79:194. RE: Erythrocyte autoantibodies in paediatric patients with
124. Cox KO, Keast D: Erythrocyte autoantibodies induced in mice sickle cell disease receiving transfusion therapy: Frequency,
immunized with rat erythrocytes. Immunology 1973;25:531–539. characteristics and significance. Br J Haematol 1999;
125. Cox KO, Keast D: Autoimmune haemolytic anaemia induced 104:189–194.
in mice immunized with rat erythrocytes. Clin Exp Immunol 148. Singer ST, Wu V, Mignacca R, Kuypers FA, Morel P, Vichinsky
1974;17:319–327. EP: Alloimmunization and erythrocyte autoimmunization in
126. Naysmith JD, Ortega-Pierres MG, Elson CJ: Rat erythrocyte- transfusion-dependent thalassemia patients of predominantly
induced anti-erythrocyte autoantibody production and asian descent. Blood 2000;96:3369–3373.
control in normal mice. Immunol Rev 1981;55:55–87. 149. Aygun B, Padmanabhan S, Paley C, Chandrasekaran V:
127. Gengozian N, McLaughlin CL: Actively induced platelet- Clinical significance of RBC alloantibodies and autoantibodies
bound IgG associated with thrombocytopenia in the mar- in sickle cell patients who received transfusions. Transfusion
moset. Blood 1978;51:1197–1210. 2002;42:37–43.
128. Gengozian N, Annostby D: Antibodies selectively reactive to 150. Kaminski ER, Hows JM, Goldman JM, Batchelor JR:
autologous and host-type platelets are obtained following Lymphocytes from multi-transfused patients exhibit cytotoxi-
interspecies immunizations in marmosets. Clin Exp Immunol city against autologous cells. Br J Haematol 1992;81:23–26.
1981;43:128–134. 151. Paglieroni TG, Ward J, Holland PV: Changes in peripheral
129. Dameshek W, Levine P: Isoimmunization with Rh factor in blood CD5 (Bla) B-cell populations and autoantibodies fol-
acquired hemolytic anemia. N Engl J Med 1943;228:641–644. lowing blood transfusion. Transfusion 1995;35:189–198.
130. Allen FH: Proceedings of the 8th Congress of the International 152. Marks JD, Ouwehand WH, Bye JM, et al: Human antibody frag-
Society of Blood Transfusion 1960:359. ments specific for human blood group antigens from a phage
131. Polesky HF, Bove JR: A fatal hemolytic transfusion reaction display library. Biotechnology (N Y): 1993;11:1145–1149.
with acute autohemolysis. Transfusion 1964;4:285–292. 153. Ouwehand WH, Bye JM, Gorick BD, et al: The humoral
132. Fudenberg HH, Rosenfield RE, Wasserman LR: Unusual immune response against blood group antigens at the molec-
specificity of autoantibody in autoimmune hemolytic anemia. ular level. Vox Sang 1994;67(Suppl 3):7–12.
J Mt Sinai Hosp 1958;25:324. 154. Issitt PD, Zellner DC, Rolih SD, Duckett JB: Autoantibodies
133. Chown B, Kaita H, Lowen B, Lewis M: Transient production of mimicking alloantibodies. Transfusion 1977;17:531–538.
anti-LW by LW-positive people. Transfusion 1971;11:220–222. 155. Issitt PD, Pavone BG: Critical re-examination of the specificity
134. Cook IA: Primary rhesus immunization of male volunteers. of auto-anti-Rh antibodies in patients with a positive direct
Brit J Haemat 1971;20:369–375. antiglobulin test. Br J Haematol 1978;38:63–74.
368 Immune Hemolytic Anemias

156. Issitt PD, Anstee DJ: Applied Blood Group Serology, 4th ed. 180. Bianchi DW: Fetal cells in the mother: From genetic diagnosis
Miami, FL: Montgomery Scientific, 1998. to diseases associated with fetal cell microchimerism. Eur J
157. Garratty G: Specificity of autoantibodies reacting optimally at Obstet Gynecol Reprod Biol 2000;92:103–108.
37°C. Immunohematology 1999;15:24–40. 181. Tyndall A, Fassas A, Passweg J, et al: Autologous haematopoi-
158. Thompson KM, Sutherland J, Barden G, et al: Human mono- etic stem cell transplants for autoimmune disease—Feasibility
clonal antibodies specific for blood group antigens demon- and transplant-related mortality. Autoimmune Disease and
strate multispecific properties characteristic of natural Lymphoma Working Parties of the European Group for Blood
autoantibodies. Immunology 1992;76:146–157. and Marrow Transplantation, the European League Against
159. Barker RN, Elson CJ: Multiple self epitopes on the Rhesus Rheumatism and the International Stem Cell Project for
polypeptides stimulate immunologically ignorant human T Autoimmune Disease. Bone Marrow Transplant 1999;24:
cells in vitro. Eur J Immunol 1994;24:1578–1582. 729–734.
160. Barker RN, Hall AM, Standen GR, Jones J, Elson CJ: 182. Johnson KL, McAlindon TE, Mulcahy E, Bianchi DW:
Identification of T-cell epitopes on the Rhesus polypeptides in Microchimerism in a female patient with systemic lupus ery-
autoimmune hemolytic anemia. Blood 1997;90:2701–2715. thematosus. Arthritis Rheum 2001;44:2107–2111.
161. Petrakis N, Politis G: Prolonged survival of viable, mitotically 183. Nelson JL: Pregnancy and microchimerism in autoimmune
competent mononuclear leukocytes in stored whole blood. disease: Protector or insurgent? Arthritis Rheum 2002;46:
New Engl J Med 1962;267:286. 291–297.
162. Turner JH, Hutchinson DL, Petricciani J: Cytogenetic and 184. Nelson JL: Microchimerism: Incidental byproduct of preg-
growth characteristics of human lymphocytes derived from nancy or active participant in human health? Trends Mol Med
stored donor blood packs. Scand J Haematol 1971;8:169–176. 2002;8:109–113.
163. McCullough J, Yunis EJ, Benson SJ, Quie PG: Effect of 185. Nelson JL: Microchimerism and human autoimmune dis-
blood-bank storage on leucocyte function. Lancet 1969; eases. Lupus 2002;11:651–654.
2:1333–1337. 186. Johnson KL, Samura O, Nelson JL, McDonnell M, Bianchi
164. Schechter GP, Soehnlen F, McFarland W: Lymphocyte DW: Significant fetal cell microchimerism in a nontransfused
response to blood transfusion in man. N Engl J Med 1972; woman with hepatitis C: Evidence of long-term survival and
287:1169–1173. expansion. Hepatology 2002;36:1295–1297.
165. Schechter GP, Whang-Peng J, McFarland W: Circulation of 187. Nelson JL: Microchimerism: Expanding new horizon in
donor lymphocytes after blood transfusion in man. Blood human health or incidental remnant of pregnancy? Lancet
1977;49:651–656. 2001;358:2011–2012.
166. Adams PT, Davenport RD, Reardon DA, Roth MS: Detection 188. Nelson JL: HLA relationships of pregnancy, microchimerism
of circulating donor white blood cells in patients receiving and autoimmune disease. J Reprod Immunol 2001;52:77–84.
multiple transfusions. Blood 1992;80:551–555. 189. Nelson JL: Microchimerism and HLA relationships of preg-
167. Houbiers JGA, Niewwenhuys C, Brand A: Chimerism after nancy: Implications for autoimmune diseases. Curr Rheu-
blood transfusion: PCR applied as a detection technique matol Rep 2001;3:222–229.
[abstract]. Vox Sanguinis 1994;67(Suppl 2):24. 190. Johnson KL, Nelson JL, Furst DE, et al: Fetal cell
168. Lee TH, Donegan E, Slichter S, Busch MP: Transient increase microchimerism in tissue from multiple sites in women with
in circulating donor leukocytes after allogeneic transfusions in systemic sclerosis. Arthritis Rheum 2001;44:1848–1854.
immunocompetent recipients compatible with donor cell pro- 191. Reed AM, Picornell YJ, Harwood A, Kredich DW: Chimerism
liferation. Blood 1995;85:1207–1214. in children with juvenile dermatomyositis. Lancet
169. Hutchinson DL, Turner JH, Schlesinger ER: Persistence of 2000;356:2156–2157.
donor cells in neonates after fetal and exchange transfusion. 192. Ishikura H, Endo J, Saito Y, et al: Graft-versus-host antibody
Am J Obstet Gynecol 1971;109:281–284. reaction causing a delayed hemolytic anemia after blood
170. Shapiro M: Familial autohemolytic anemia and runting syn- transfusion. [letter]. Blood 1993;82:3222–3223.
drome with Rh-o-specific autoantibody. Transfusion 1967; 193. Salama A, Mueller-Eckhardt C: Delayed hemolytic trans-
7:281–296. fusion reactions. Evidence for complement activation
171. Fudenberg HH, Solomon A: “Acquired agammaglobuline- involving allogeneic and autologous red cells. Transfusion
mia” with autoimmune hemolytic disease: Graft-versus-host 1984;24:188–193.
reaction? Vox Sang 1961;6:68. 194. Ness PM, Shirey RS, Thoman SK, Buck SA: The differentiation
172. Hobbs JR, Russell A, Worlledge SM: Dysgammaglobu- of delayed serologic and delayed hemolytic transfusion reac-
linaemia type IV C. Clin Exp Immunol 1967;2:589–599. tions: Incidence, long-term serologic findings, and clinical
173. Schaller J, Davis SD, Ching YC, Lagunoff D, Williams CP, significance. Transfusion 1990;30:688–693.
Wedgwood RJ: Hypergammaglobulinaemia, antibody 195. Dzik WH: Microchimerism after transfusion: The spectrum
deficiency, autoimmune haemolytic anaemia, and nephritis from GVHD to alloimmunization. Transfus Sci 1995;16:107–108.
in an infant with a familial lymphopenic immune defect. 196. Zupanska B, Lawkowicz W, Gorska B, et al: Autoimmune
Lancet 1966;2:825–829. haemolytic anaemia in children. Br J Haematol 1976;34:511–520.
174. Lee TH, Reed W, Mangawang-Montalvo L, Watson J, Busch 197. Habibi B, Homberg JC, Schaison G, Salmon C: Autoimmune
MP: Donor WBCs can persist and transiently mediate hemolytic anemia in children: A review of 80 cases. Am J Med
immunologic function in a murine transfusion model: Effects 1974;56:61–69.
of irradiation, storage, and histocompatibility. Transfusion 198. Buchanan GR, Boxer LA, Nathan DG: The acute and transient
2001;41:637–642. nature of idiopathic immune hemolytic anemia in childhood.
175. Goodarzi MO, Lee TH, Pallavicini MG, Donegan EA, Busch J Pediatr 1976;88:780–783.
MP: Unusual kinetics of white cell clearance in transfused 199. Sokol RJ, Hewitt S, Stamps BK, Hitchen PA: Autoimmune
mice. Transfusion 1995;35:145–149. haemolysis in childhood and adolescence. Acta Haematol
176. Lee TH, Paglieroni T, Ohto H, Holland PV, Busch MP: Survival 1984;72:245–257.
of donor leukocyte subpopulations in immunocompetent 200. Heddle NM: Acute paroxysmal cold hemoglobinuria.
transfusion recipients: Frequent long-term microchimerism in Transfus Med Rev 1989;3:219–229.
severe trauma patients. Blood 1999;93:3127–3139. 201. Gottsche B, Salama A, Mueller-Eckhardt C: Donath-
177. Nelson JL: Microchimerism and scleroderma. Curr Rheumatol Landsteiner autoimmune hemolytic anemia in children: A
Rep 1999;1:15–21. study of 22 cases. Vox Sang 1990;58:281–286.
178. Nelson JL: Microchimerism: Implications for autoimmune 202. Nordhagen R, Stensvold K, Winsnes A, Skyberg D, Storen A:
disease. Lupus 1999;8:370–374. Paroxysmal cold haemoglobinuria: The most frequent acute
179. Famularo G, De Simone C: Systemic sclerosis from autoim- autoimmune haemolytic anaemia in children? Acta Paediatr
munity to alloimmunity. South Med J 1999;92:472–476. Scand 1984;73:258–262.
Unusual Aspects of Acquired Immune Hemolytic Anemias 369

203. Gurgey A, Yenicesu I, Kanra T, et al: Autoimmune hemolytic 227. Johnson CA, Abildgaard CF: Treatment of idiopathic autoim-
anemia with warm antibodies in children: Retrospective mune hemolytic anemia in children. Review and report of
analysis of 51 cases. Turk J Pediatr 1999;41:467–471. two fatal cases in infancy. Acta Paediatr Scand 1976;
204. Heisel MA, Ortega JA: Factors influencing prognosis in child- 65:375–379.
hood autoimmune hemolytic anemia. Am J Pediatr Hematol 228. Sasaki H, Akutagawa H, Kuwakado K, Uemura M, Emi I:
Oncol 1983;5:147–152. High-dose intravenous IgG therapy in a seven-week-old
205. Carapella de Luca E, Casadei AM, di Piero G, Midulla M, infant with chronic autoimmune hemolytic anemia. Am J
Bisdomini C, Purpura M: Auto-immune haemolytic anaemia Hematol 1987;25:215–218.
in childhood: Follow-up in 29 cases. Vox Sang 1979;36:13–20. 229. Otheo E, Maldonado MS, Munoz A, Hernandez-Jodra M:
206. Ammann AJ, Hong R: Selective IgA deficiency: Presentation High-dose intravenous immunoglobulin as single therapy in
of 30 cases and a review of the literature. Medicine (Baltimore) a child with autoimmune hemolytic anemia. Pediatr Hematol
1971;50:223–236. Oncol 1997;14:487–490.
207. Miescher PA, Tucci A, Beris P, Favre H: Autoimmune 230. McConnell ME, Atchison JA, Kohaut E, Castleberry RP:
hemolytic anemia and/or thrombocytopenia associated with Successful use of plasma exchange in a child with refractory
lupus parameters. Semin Hematol 1992;29:13–17. immune hemolytic anemia. Am J Pediatr Hematol Oncol
208. Kalmanti M, Polychronopoulou S: Autoimmune hemolytic 1987;9:158–160.
anemia as an initial symptom in childhood Hodgkin’s disease. 231. Poschmann A, Fischer K: Autoimmune haemolytic anaemia:
Pediatr Hematol Oncol 1992;9:393–395. Recent advances in pathogenesis, diagnosis and treatment.
209. Duru F, Gurgey A, Cetin M, Kanra T, Altay C: Chronic autoim- Eur J Pediatr 1985;143:258–260.
mune hemolytic anemia in children: A report of four patients. 232. Swisher SN: Acquired hemolytic disease. Postgrad Med
J Med 1994;25:231–240. 1966;40:378–386.
210. Perez-Atayde AR, Sirlin SM, Jonas M: Coombs-positive 233. Benraad CE, Scheerder HA, Overbeeke MA: Autoimmune
autoimmune hemolytic anemia and postinfantile giant cell haemolytic anaemia during pregnancy. Eur J Obstet Gynecol
hepatitis in children. Pediatr Pathol 1994;14:69–77. Reprod Biol 1994;55:209–211.
211. Bernard O, Hadchouel M, Scotto J, Odievre M, Alagille D: 234. Sokol RJ, Hewitt S, Stamps BK: Erythrocyte autoantibodies, auto-
Severe giant cell hepatitis with autoimmune hemolytic immune haemolysis and pregnancy. Vox Sang 1982;43:169–176.
anemia in early childhood. J Pediatr 1981;99:704–711. 235. Hoppe B, Stibbe W, Bielefeld A, Pruss A, Salama A: Increased
212. Miyazaki S, Nakayama K, Akabane T, et al: Follow-up study RBC autoantibody production in pregnancy. Transfusion
of 34 children with autoimmune hemolytic anemia. Nippon 2001;41:1559–1561.
Ketsueki Gakkai Zasshi 1983;46:6–10. 236. Buyon JP: The effects of pregnancy on autoimmune diseases.
213. Olcay L, Duzova A, Gumruk F: A warm antibody mediated J Leukoc Biol 1998;63:281–287.
acute hemolytic anemia with reticulocytopenia in a four- 237. Chaplin H, Jr, Cohen R, Bloomberg G, Kaplan HJ, Moore JA,
month-old girl requiring immunosuppressive therapy. Turk J Dorner I: Pregnancy and idiopathic autoimmune haemolytic
Pediatr 1999;41:239–244. anaemia: A prospective study during 6 months gestation and
214. Liesveld JL, Rowe JM, Lichtman MA: Variability of the ery- 3 months post-partum. Brit J Haemat 1973;24:219–229.
thropoietic response in autoimmune hemolytic anemia: 238. Quinlivan WL, Goldberg S: Autoimmune hemolytic anemia of
Analysis of 109 cases. Blood 1987;69:820–826. the cold antibody type associated with pregnancy. Am J
215. Zuelzer WW, Mastrangelo R, Stulberg CS, Poulik MD, Page Obstet Gynecol 1967;98:1102–1104.
RH, Thompson RI: Autoimmune hemolytic anemia. Natural 239. Jain S, Agarwal S, Dash SC, Grewal KS: Autoimmune
history and viral-immunologic interactions in childhood. Am hemolytic anaemia and foetal loss. J Assoc Physicians India
J Med 1970;49:80–93. 1977;25:765–766.
216. Friedmann AM, King KE, Shirey RS, Resar LM, Casella JF: 240. Issaragrisil S, Kruatrachue M: An association of pregnancy
Fatal autoimmune hemolytic anemia in a child due to warm- and autoimmune haemolytic anaemia. Scand J Haematol
reactive immunoglobulin M antibody. J Pediatr Hematol 1983;31:63–68.
Oncol 1998;20:502–505. 241. Lawe JE: Successful exchange transfusion of an infant for
217. McCann EL, Shirey RS, Kickler TS, Ness PM: IgM autoagglu- AIHA developing late in mother’s pregnancy. Transfusion
tinins in warm autoimmune hemolytic anemia: A poor prog- 1982;22:66–68.
nostic feature. Acta Haematol 1992;88:120–125. 242. Baumann R, Rubin H: Autoimmune hemolytic anemia during
218. Shirey RS, Kickler TS, Bell W, Little B, Smith B, Ness PM: Fatal pregnancy with hemolytic disease in the newborn. Blood
immune hemolytic anemia and hepatic failure associated with 1973;41:293–297.
a warm-reacting IgM autoantibody. Vox Sang 1987;52:219–222. 243. Vedovini F, Benedetti PA: Anemia emolitica da autoanticorpi
219. Waterbury L, Parnes H, Katz RS: Fatal warm antibody materni. Riv Clin Pediatr 1963;72:339.
autoimmune hemolysis with marked erythrocyte autoaggluti- 244. Burt RL, Prichard RW: Acquired hemolytic anemia in preg-
nation and liver necrosis. South Med J 1986;79:646–647. nancy: Report of a case. Obstet Gynecol 1957;90:444.
220. Freedman J, Wright J, Lim FC, Garvey MB: Hemolytic warm 245. Soderhjelm L: Non-spherocytic haemolytic anaemia in mother
IgM autoagglutinins in autoimmune hemolytic anemia. and new-born infant. Acta Paediatrica 1959;48(Suppl. 117):34.
Transfusion 1987;27:464–467. 246. Seip M: Systemic lupus erythematosus in pregnancy with
221. Win N, Kaye T, Mir N, Damain-Willems C, Chatfield C: haemolytic anaemia, leucopenia and thrombocytopenia in the
Autoimmune haemolytic anaemia in infancy with anti-Kpb mother and her newborn infant. Arch Dis Child 1960;35:364.
specificity. Vox Sang 1996;71:187–188. 247. Letts HW, Kredentser B: Thrombocytopenia, hemolytic
222. Ritz ND, Haber A: Autoimmune hemolytic anemia in a 6 anemia, and two pregnancies: Report of a case. Am J Clin
week old child. J Pediatr 1962;61:904–910. Pathol 1968;49:481–486.
223. Oski FA, Abelson NM: Autoimmune hemolytic anemia in an 248. Silverstein MN, Aaro LA, Kempers RD: Evans’ syndrome and
infant. Report of a case treated unsuccessfully with thymec- pregnancy. Am J Med Sci 1966;252:206–211.
tomy. J Pediatr 1965;67:752–758. 249. Passi GR, Kriplani A, Pati HP, Choudhry VP: Isoimmune
224. Bakx CJA, van Loghem JJ: Acquired hemolytic anemia in a hemolysis in an infant due to maternal Evans’ syndrome.
newborn. Vox Sang 1953;42:79. Indian J Pediatr 1997;64:893–895.
225. Erler BS, Smith L, McQuiston D, Pepkowitz SH, Goldfinger D: 250. Sacks DA, Platt LD, Johnson CS: Autoimmune hemolytic disease
Red cell autoantibody production in utero: A case report. during pregnancy. Am J Obstet Gynecol 1981;140:942–946.
Transfusion 1994;34:72–74. 251. Yam P, Wilkinson L, Petz LD, Garratty G: Studies on
226. Hadnagy C: Severe chronic autoimmune haemolytic anaemia hemolytic anemia in pregnancy with evidence for autoimmu-
presenting haemolytic disease of the newborn. Lancet 1989; nization in a patient with a negative direct antiglobulin
ii:749. (Coombs’) test. Am J Hematol 1980;8:23–29.
370 Immune Hemolytic Anemias

252. Ng SC, Wong KK, Raman S, Bosco J: Autoimmune haemolytic 275. Gottlieb P, Axelsson O, Bakos O, Rastad J: Splenectomy
anaemia in pregnancy: A case report. Eur J Obstet Gynecol during pregnancy: An option in the treatment of autoimmune
Reprod Biol 1990;37:83–85. thrombocytopenic purpura. Br J Obstet Gynaecol 1999;106:
253. Tsai YC, Chang JM, Chang JC, Changahien CC, Chen PH, Jeng 373–375.
TT: Idiopathic autoimmune hemolytic anemia during preg- 276. Sendag F, Kazandi M, Terek MC: Splenectomy combined with
nancy. J Formos Med Assoc 1994;93:328–331. cesarean section in a patient with severe immunological
254. de Groot AW: A pregnant woman with hemolytic anemia [in thrombocytopenic purpura refractory to medical therapy.
Dutch]. Ned Tijdschr Verloskd Gynaecol 1969;69:283–286. J Obstet Gynaecol Res 2001;27:85–88.
255. Eldor A, Yatziv S, Hershko C: Relapsing Coombs-negative 277. Iwase K, Higaki J, Yoon HE, et al: Hand-assisted laparoscopic
haemolytic anaemia in pregnancy with haemolytic disease in splenectomy for idiopathic thrombocytopenic purpura during
the newborn. Br Med J 1975;4:625. pregnancy. Surg Laparosc Endosc Percutan Tech 2001;11:53–56.
256. Hershko C, Berrebi A, Resnitzky P, Eldor A: Relapsing 278. Burrows R: Splenectomy during pregnancy: An option in
haemolytic anaemia of pregnancy with negative antiglobulin treatment of autoimmune thrombocytopenic purpura. Br J
reaction. Scand J Haematol 1976;16:135–140. Obstet Gynaecol 1999;106:1330–1331.
257. Goodall HB, Ho-Yen DO, Clark DM, Thomson MA, Browning 279. Hardwick RH, Slade RR, Smith PA, Thompson MH:
MC, Crowder AM: Haemolytic anaemia of pregnancy. Scand J Laparoscopic splenectomy in pregnancy. J Laparoendosc Adv
Haematol 1979;22:185–191. Surg Tech A 1999;9:439–440.
258. Starksen NF, Bell WR, Kickler TS: Unexplained hemolytic 280. Kissmeyer-Nielsen FS, Grent-Hansen K, Kieler J: Immuno-
anemia associated with pregnancy. Am J Obstet Gynecol hemolytic anemia with familial occurrence. Acta Med Scand
1983;146:617–622. 1952;144:35–39.
259. Bjornsson S, Brennand JE, Calder AA, et al: Unexplained 281. Fialkow PJ, Fudenberg HH, Epstein WV: “Acquired” anti-
haemolytic anaemia in successive pregnancies with negative body hemolytic anemia and familial aberrations in gamma
direct antiglobulin test and response to high dose i.v. IgG. Br J globulins. Amer J Med 1964;36:188–199.
Obstet Gynaecol 1994;101:75–77. 282. Olanoff LS, Fudenberg HH: Familial autoimmunity: Twenty
260. Kumar R, Advani AR, Sharan J, Basharutallah MS, Al Lumai years later. J Clin Lab Immunol 1983;11:105–111.
AS: Pregnancy induced hemolytic anemia: An unexplained 283. Hennemann HH, Krause H: Chronische Thrombozytopenie
entity. Ann Hematol 2001;80:623–626. (Morbus Werlhof) und erworbene hamolytische Anamie bei
261. Craig GA, Turner RL: A case of symptomatic haemolytic zwei Schwestern. Dtsch med Wschr 1964;89:1161–1166.
anaemia in pregnancy. Br Med J 1955;i:1003–1005. 284. Dobbs CE: Familial auto-immune hemolytic anemia. Arch
262. Jankelowitz T, Eckerling B, Joshua H: A case of acquired Intern Med 1965;116:273–276.
haemolytic anaemia associated with pregnancy. S Afr Med J 285. Loghem-Langereis E, Peetoom F, van der HM, Van Loghem JJ,
1976;34:911–913. Bosch E, Goudsmit R: The occurrence of gammaglobulin/
263. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Warm- anti-gammaglobulin complexes in a patient suffering from
antibody syndromes I: “idiopathic” types: history and clinical hypogammaglobulinaemia and haemolytic anaemia. Bibl
features. In The Haemolytic Anaemias, 3rd ed.,Vol. 3, The Haematol 1965;23:55–61.
Auto-Immune Haemolytic Anaemias. New York: Churchill 286. Cordova MS, Baez-Villasenor J, Mendez JJ, Campos E:
Livingstone, 1992:6–53. Acquired hemolytic anemia with positive antiglobulin
264. Bermas BL, Hill JA: Effects of immunosuppressive drugs Coombs’ test) in mother and daughter. Arch Intern Med
during pregnancy. Arthritis Rheum 1995;38:1722–1732. 1966;117:692–695.
265. Janssen NM, Genta MS: The effects of immunosuppressive 287. Schwartz RS, Costea N: Autoimmune hemolytic anemia:
and anti-inflammatory medications on fertility, pregnancy, Clinical correlations and biological implications. Semin
and lactation. Arch Intern Med 2000;160:610–619. Hematol 1966;3:2–26.
266. Schatz M, Patterson R, Zeitz S, O’Rourke J, Melam H: 288. Shapiro M: Familial autohemolytic anemia and runting
Corticosteroid therapy for the pregnant asthmatic patient. syndrome with Rh-o-specific autoantibody. Transfusion
JAMA 1975;233:804–807. 1967;7:281–296.
267. NIH Consensus Development Panel on the Effect of 289. Pirofsky B: Autoimmunization and the Autoimmune
Corticosteroids for Fetal Maturation on Perinatal Outcomes. Hemolytic Anemias. Baltimore, Md: Williams & Wilkins, 1969.
Effect of corticosteroids for fetal maturation on perinatal out- 290. Pirofsky B: Hereditary aspects of autoimmune hemolytic
comes. JAMA 1995;273:413–418. anemia; a retrospective analysis. Vox Sang 1968;14:334–347.
268. Armenti VT, Ahlswede KM, Ahlswede BA, Jarrell BE, Moritz 291. Seip M, Harboe M, Cyvin K: Chronic autoimmune hemolytic
MJ, Burke JF: National transplantation Pregnancy Registry— anemia in childhood with cold antibodies, aplastic crises, and
Outcomes of 154 pregnancies in cyclosporine-treated female familial occurrence. Acta Paediatr Scand 1969;58:275–280.
kidney transplant recipients. Transplantation 1994;57:502–506. 292. Pollock JG, Fenton E, Barrett KE: Familial autoimmune
269. Haugen G, Fauchald P, Sodal G, Leivestad T, Moe N: Pregnancy haemolytic anaemia associated with rheumatoid arthritis and
outcome in renal allograft recipients in Norway: The importance pernicious anaemia. Br J Haematol 1970;18:171–182.
of immunosuppressive drug regimen and health status before 293. Zuelzer WW, Mastrangelo R, Stulberg CS, Poulik MD, Page
pregnancy. Acta Obstet Gynecol Scand 1994;73:541–546. RH, Thompson RI: Autoimmune hemolytic anemia. Natural
270. Shaheen FA, al-Sulaiman MH, al-Khader AA: Long-term history and viral-immunologic interactions in childhood. Am
nephrotoxicity after exposure to cyclosporine in utero. J Med 1970;49:80–93.
Transplantation 1993;56:224–225. 294. Blajchman MA, Hui YT, Jopnes TE, Luke KH: Familial
271. Potter M, Stockley R, Storry J, Slade R: ABO alloimmunisation autoimmune hemolytic anemia with an autoantibody
after intravenous immunoglobulin infusion. Lancet 1988; demonstrating U specificity [abstract]. Program of the 24th
1:932–933. Annual Meeting of the American Association of Blood Banks,
272. Letsky EA, Warwick R: Hematological Problems. In James PK, 1971:82.
Steer PJ, Weiner CP, Gonik B (eds): High Risk Pregnancy: 295. Dacie JV, Worlledge SM: Auto-allergic blood diseases. In Gell
Management Options. London: WB Saunders, 1996:359–372. PGH, Coombs RRA, Lachmann PJ (eds): Clinical Aspects of
273. Sipes SL, Weiner C: Coagulation Disorders in Pregnancy. In Immunology. Oxford: Blackwell Scientific, 1975:1149–1182.
Reece EA, Hobbins JC, Mahoney MJ, Petrie RH (eds): 296. Roth P, Morell A, Hunziker HR, Gehri P, Bucher U: Familial
Medicine of the Fetus and Mother. Philadelphia: JB autoimmune hemolytic animia (AIHA) with negative Coombs
Lippincott, 1992:1111–1138. test, lymphocytopenia and hypogammaglobulinemia [in
274. Letsky EA: Coagulation Defects. In de Swiet M (ed): Medical German]. Schweiz Med Wochenschr 1975;105:1584–1585.
Disorders in Obstetric Practice. Oxford: Blackwell Science, 297. Toolis F, Parker AC, White A, Urbaniak S: Familial autoim-
1989:104–165. mune haemolytic anaemia. Br Med J 1977;1:1392.
Unusual Aspects of Acquired Immune Hemolytic Anemias 371

298. Conley CL, Savarese DM: Biologic false-positive serologic 322. Colmers RA, Snavely JG: Acute hemolytic anemia in primary
tests for syphilis and other serologic abnormalities in autoim- atypical pneumonia produced by exposure and chilling.
mune hemolytic anemia and thrombocytopenic purpura. N Engl J Med 1947;237:505–510.
Medicine (Baltimore) 1989;68:67–84. 323. Dake SB, Johnston MF, Brueggeman P, Barner HB: Detection
299. Reynolds MV, Vengelen-Tyler V, Morel PA: Autoimmune of cold hemagglutination in a blood cardioplegia unit before
hemolytic anemia associated with autoanti-Ge. Vox Sang systemic cooling of a patient with unsuspected cold agglu-
1981;41:61–67. tinin disease. Ann Thorac Surg 1989;47:914–915.
300. Perez-Mateo M, Tascon A: Idiopathic autoimmune hemolytic 324. Schmidt PJ: Cold agglutinins and hypothermia [letter]. Arch
anemia in twins [in Spanish]. Med Clin (Barc ) 1982;79:476. Intern Med 1985;145:578–579.
301. Boling EP, Wen J, Reveille JD, Bias WB, Chused TM, Arnett FC: 325. Agarwal SK, Ghosh PK, Gupta D: Cardiac surgery and cold-
Primary Sjogren’s syndrome and autoimmune hemolytic reactive proteins. Ann Thorac Surg 1995;60:1143–1150.
anemia in sisters: A family study. Am J Med 1983;74:1066–1071. 326. Moore RA, Geller EA, Mathews ES, Botros SB, Jose AB, Clark
302. Horowitz SD, Borcherding W, Hong R: Autoimmune DL: The effect of hypothermic cardiopulmonary bypass on
hemolytic anemia as a manifestation of T-suppressor-cell patients with low- titer, nonspecific cold agglutinins. Ann
deficiency. Clin Immunol Immunopathol 1984;33:313–323. Thorac Surg 1984;37:233–238.
303. McLeod AG, Pai M, Carter RF, Squire J, Barr RD: Familial 327. Nydegger U, Hardegger T, Tobler A, Rieder H, Cerniak A,
Evans syndrome: A report of an affected sibship. J Pediatr Lammle B: Cold agglutinin syndrome and liver transplanta-
Hematol Oncol 1999;21:244–247. tion. Vox Sang 1994;67:85.
304. Lippman SM, Arnett FC, Conley CL, Ness PM, Meyers DA, 328. Klein HG, Faltz LL, McIntosh CL, Appelbaum FR, Deisseroth
Bias WB: Genetic factors predisposing to autoimmune AB, Holland PV: Surgical hypothermia in a patient with a cold
diseases: Autoimmune hemolytic anemia, chronic thrombocy- agglutinin: Management by plasma exchange. Transfusion
topenic purpura, and systemic lupus erythematosus. Am J 1980;20:354–357.
Med 1982;73:827–840. 329. Williams AC: Cold agglutinins: Cause for concern? Anaes-
305. Schmid FR: Pirofsky, editor. 1969 [personal communication]. thesia 1980;35:887–889.
306. Habibi B, Homberg JC, Schaison G, Salmon C: Autoimmune 330. Berreklouw E, Moulijn AC, Pegels JG, Meijne NG: Myocardial
hemolytic anemia in children: A review of 80 cases. Am J Med protection with cold cardioplegia in a patient with cold autoag-
1974;56:61–69. glutinins and hemolysins. Ann Thorac Surg 1982;33:521–522.
307. Conley CL: Immunologic precursors of autoimmune hema- 331. Blumberg N, Hicks G, Woll J, et al: Successful cardiac bypass
tologic disorders: Autoimmune hematologic disorders: surgery in the presence of a potent cold agglutinin without
Autoimmune hemolytic anemia and thrombocytopenic plasma exchange [letter]. Transfusion 1983;23:363.
purpura. Johns Hopkins Med J 1981;149:101–109. 332. Diaz JH, Cooper ES, Ochsner JL: Cold hemagglutination
308. Shahian DM, Wallach SR, Bern MM: Open heart surgery in pathophysiology: Evaluation and management of patients
patients with cold-reactive proteins. Surg Clin North Am undergoing cardiac surgery with induced hypothermia. Arch
1985;65:315–322. Intern Med 1983;144:1639–1641.
309. Park JV, Weiss CI: Cardiopulmonary bypass and myocardial 333. Landymore R: Cold agglutinins and coronary artery bypass
protection: Management problems in cardiac surgical patients grafting. Ann Thorac Surg 1983;35:472.
with cold autoimmune disease [review]. Anesth Analg 334. Landymore R, Isom W, Barlam B: Management of patients
1988;67:75–78. with cold agglutinins who require open-heart surgery. Can J
310. Menasche P, Subayi JB, Piwnica A: Retrograde coronary sinus Surg 1983;26:79–80.
cardioplegia for aortic valve operations: A clinical report on 335. Leach AB, Van Hasselt GL, Edwards JC: Cold agglutinins and
500 patients. Ann Thorac Surg 1990;49:556–563. deep hypothermia. Anaesthesia 1983;38:140–143.
311. Izzat MB, Rajesh PB, Smith GH: Use of retrograde cold crys- 336. Andrzejewski C, Jr, Gault E, Briggs M, Silberstein L: Benefit of
talloid cardioplegia in a patient with unexpected cold aggluti- a 37 degree C extracorporeal circuit in plasma exchange
nation. Ann Thorac Surg 1993;56:1395–1397. therapy for selected cases with cold agglutinin disease. J Clin
312. Shulman IA, Petz LD: Red cell compatibility testing: Clinical Apheresis 1988;4:13–17.
significance and laboratory methods. In Petz LD, Swisher SN, 337. Paccagnella A, Simini G, Nieri A, Da Col U, Frugoni C, Valfre
Kleinman S, Spence RK, Strauss RG (eds): Clinical Practice of C: Cardiopulmonary bypass and cold agglutinin. J Thorac
Transfusion Medicine, 3rd ed. New York: Churchill Cardiovasc Surg 1988;95:543.
Livingstone, 1996:199–244. 338. Lee MC, Chang CH, Hsieh MJ: Use of a total wash-out method
313. Bedrosian CL, Simel DL: Cold hemagglutinin disease in the in an open heart operation. Ann Thorac Surg 1989;47:57–58.
operating room. South Med J 1987;80:466–471. 339. Aoki A, Kay GL, Zubiate P, Ruggio J, Kay JH: Cardiac opera-
314. Wertlake PT, McGinniss MH, Schmidt PJ: Cold antibody and tion without hypothermia for the patient with cold agglutinin.
persistent intravascular hemolysis after surgery under Chest 1993;104:1627–1629.
hypothermia. Transfusion 1969;9(2):70–73. 340. Beebe DS, Bergen L, Palahniuk RJ: Anesthetic management of
315. Diaz JH, Cooper ES, Ochsner JL: Cardiac surgery in patients a patient with severe cold agglutinin hemolytic anemia utiliz-
with cold autoimmune diseases. Anesth Analg 1984;63:349–352. ing forced air warming. Anesth Analg 1993;76:1144–1146.
316. Bracken CA, Gurkowski MA, Naples JJ, et al: Case 6—1993. 341. Hearnsberger J, Ziomek S, Tobler G, Maxson T, VanDevanter
Cardiopulmonary bypass in two patients with previously S, Harrell JE, Jr: Management of cold agglutinemia with warm
undetected cold agglutinins. J Cardiothorac Vasc Anesth heart surgical intervention: A case report. J Thorac Cardiovasc
1993;7:743–749. Surg 1993;106:756–757.
317. Holman WL, Smith SH, Edwards R, Huang ST: Agglutination 342. Zoppi M, Oppliger R, Althaus U, Nydegger U: Reduction of
of blood cardioplegia by cold-reacting autoantibodies. Ann plasma cold agglutinin titers by means of plasmapheresis to
Thorac Surg 1991;51:833–835. prepare a patient for coronary bypass surgery. Infusionsther
318. Stalker AL: Intravascular erythrocyte aggregation. Bibl Anat Transfusionsmed 1993;20:19–22.
1964;4:108–111. 343. Mastrogiovanni G, Masiello P, Iesu S, Senese I, Di Benedetto
319. Davidsohn I, Lee CL, Takahashi T: Hepatic infarcts in mice G: Management of cold agglutinemia with intermittent warm
injected with anti-erythrocytic serum. Arch Pathol 1963;76: blood cardioplegia and normothermia. Ann Thorac Surg 1996;
398–403. 62:317.
320. AuBuchon JP, Scofan BA, Davey RJ: Hemolysis during extracor- 344. Onoe M, Magara T, Yamamoto Y: Cardiac operation for a
poreal circulation: Signifiance of cold-reactive auto-antibodies patient with autoimmune hemolytic anemia with warm-
and mechanical trauma [abstract]. Blood 1983;65:42a. reactive antibodies. Ann Thorac Surg 2001;71:351–352.
321. Niejadlik DC, Lozner EL: Cooling mattress induced acute 345. Ko W, Isom OW: Cardiopulmonary bypass procedures in
hemolytic anemia. Transfusion 1974;14:145–147. patients with cold-reactive hemagglutination: A case report
372 Immune Hemolytic Anemias

and a literature review. J Cardiovasc Surg (Torino) 1996;37: 372. Branch DR, Hian AL, Carlson F, Maslow WC, Petz LD:
623–626. Erythrocyte age-fractionation using a Percoll-Renografin
346. Lichtenstein SV, Salerno TA, Slutsky AS: Pro: Warm continu- density gradient: Application to autologous red cell antigen
ous cardioplegia is preferable to intermittent hypothermic determinations in recently transfused patients. Am J Clin
cardioplegia for myocardial protection during cardiopul- Pathol 1983;80:453–458.
monary bypass. J Cardiothorac Anesth 1990;4:279–281. 373. Technical Manual, 14th ed. Brecher, M (ed). Bethesda, Md:
347. Salerno TA, Houck JP, Barrozo CA, et al: Retrograde continu- American Association of Blood Banks, 2002.
ous warm blood cardioplegia: A new concept in myocardial 374. Reid ME, Toy PT: Simplified method for recovery of autolo-
protection. Ann Thorac Surg 1991;51:245–247. gous red blood cells from transfused patients. Am J Clin
348. Lichtenstein SV, Ashe KA, el Dalati H, Cusimano RJ, Panos A, Pathol 1983;79:364–366.
Slutsky AS: Warm heart surgery. J Thorac Cardiovasc Surg 375. Vengelen-Tyler V, Gonzales B: Reticulocyte rich RBCs will
1991;101:269–274. give weak reactions with many blood typing antisera
349. Roe BB: Warm blood cardioplegia: Back to square one. Ann [abstract]. Transfusion 1985;25:476.
Thorac Surg 1993;55:330–331. 376. Garratty G, Arndt PA: Applications of flow cytofluorometry to
350. Muehrcke DD, Torchiana DF: Warm heart surgery in patients red blood cell immunology. Cytometry 1999;38:259–267.
with cold autoimmune disorders. Ann Thorac Surg 1993; 377. Griffin GD, Lippert LE, Dow NS, Berger TA, Hickman MR,
55:532–533. Salata KF: A flow cytometric method for phenotyping recipient
351. Gokhale AG, Suhasini T, Saraswati V, Chandrasekhar N, red cells following transfusion. Transfusion 1994;34:233–237.
Rajagopal P: Cold agglutinins and warm heart surgery. 378. Wagner F: Identification of recipient Rh phenotype in a chron-
J Thorac Cardiovasc Surg 1993;105:557. ically transfused child by two-colour immunofluorescence.
352. Donatelli F, Mariani MA, Triggiani M, Pocar M, Santoro F, Transfus Med 1994;4:205–208.
Grossi A: Warm heart surgery in cold haemagglutinin disease. 379. Wenk RE, Chiafari PA: DNA typing of recipient blood after
Cardiovasc Surg 1995;3:191–192. massive transfusion. Transfusion 1997;37:1108–1110.
353. Taft EG, Propp RP, Sullivan SA: Plasma exchange for cold 380. Reid ME, Rios M, Powell VI, Charles-Pierre D, Malavade V:
agglutinin hemolytic anemia. Transfusion 1977;17:173–176. DNA from blood samples can be used to genotype patients
354. Rodenhuis S, Maas A, Hazenberg CA, Das PC, Nieweg HO: who have recently received a transfusion. Transfusion
Inefficacy of plasma exchange in cold agglutinin hemolytic 2000;40:48–53.
anemia—A case study. Vox Sang 1985;49:20–25. 381. Castilho L, Rios M, Pellegrino J, Rodrigues A, Costa FF: Blood
355. Shumak KH, Rock GA: Therapeutic plasma exchange. N Engl group genotyping for the management of patients with
J Med 1984;310:762–771. “warm” antibody-induced hemolytic anemia (WAIHA)
356. Pineda AA, Brzica SM, Jr, Taswell HF: Hemolytic transfusion [abstract]. Blood 2001;98:62a.
reaction. Recent experience in a large blood bank. Mayo Clin 382. Petz LD: The expanding boundaries of transfusion medicine.
Proc 1978;53:378–390. In Nance SJ (ed): Clinical and Basic Science Aspects of
357. Myhre BA: Fatalities from blood transfusion. JAMA Immunohematology. Arlington, VA: American Association of
1980;244:1333–1335. Blood Banks, 1991:73–113.
358. Honig CL, Bove JR: Transfusion-associated fatalities: Review 383. Petz LD: Blood transfusion in hemolytic anemias. Immuno-
of Bureau of Biologics reports 1976–1978. Transfusion hematology 1999;15:15–23.
1980;20:653–661. 384. Dameshek W: Autoimmunity: Theoretical aspects. Ann N Y
359. Sazama K: Reports of 355 transfusion-associated deaths: 1976 Acad Sci 1965;124:6–28.
through 1985. Transfusion 1990;30:583–590. 385. Garratty G: Novel Mechanisms for Immune Destruction of
360. Linden JV, Kaplan HS: Transfusion errors: Causes and effects. Circulating Autologous Cells. In Silberstein LE (ed):
[review]. Transfus Med Rev 1994;8:169–183. Autoimmune Disorders of Blood. Bethesda, Md: American
361. Williamson L, Cohen H, Love E, Jones H, Todd A, Soldan K: The Association of Blood Banks, 1996:79–114.
Serious Hazards of Transfusion (SHOT) initiative: The UK 386. Thompson RA, Rowe DS: Reactive haemolysis—A distinctive
approach to haemovigilance. Vox Sang 2000;78(Suppl. 2): form of red cell lysis. Immunology 1968;14:745–762.
291–295. 387. Thompson RA, Lachmann PJ: Reactive lysis: The comple-
362. Croucher BEE: Differential diagnosis of delayed hemolytic ment-mediated lysis of unsensitized cells. I: The characteriza-
transfusion reaction. In Bell CA (ed): A Seminar on Laboratory tion of the indicator factor and its identification as C7. J Exp
Management of Hemolysis. Washington, DC: American Med 1970;131:629–641.
Association of Blood Banks, 1979:151–160. 388. Lachmann PJ, Thompson RA: Reactive lysis: The comple-
363. Blood Transfusion in Clinical Medicine, 10th ed., Mollison PL, ment-mediated lysis of unsensitized cells. II: The characteri-
Engelfriet CP, Contreras M (ed). Oxford: Blackwell Science, 1997. zation of activated reactor as C56 and the participation of C8
364. Holland PV, Wallerstein RO: Delayed hemolytic transfusion and C9. J Exp Med 1970;131:643–657.
reaction with acute renal failure. JAMA 1968;204:1007–1008. 389. Götze O, Muller-Eberhard HJ: Lysis of erythrocytes by comple-
365. Croucher BEE, Crookston MC, Crookston JH: Delayed ment in the absence of antibody. J Exp Med 1970;132:898–915.
haemolytic transfusion reactions simulating auto-immune 390. Goldman JN, Ruddy S, Austen KF: Reaction mechanisms of
haemolytic anaemia. Vox Sang 1967;12:32. nascent C567 (reactive lysis). I: Reaction characteristics for
366. Thomson S, Johnstone M: Delayed haemolytic transfusion production of EC567 and lysis by C8 and C9. J Immunol
reaction due to anti Jk a and anti M. Can J Med Technol 1972;109:353–359.
1972;34:159–161. 391. McLeod BC, Baker P, Gewurz H. Studies on the inhibition of
367. Rothman IK, Alter HJ, Strewler GJ: Delayed overt hemolytic C56-initiated lysis (reactive lysis). II: C567-INH—An inhibitor
transfusion reaction due to anti-U antibody. Transfusion of the C567 trimolecular complex of complement. Int Arch
1976;16:357–360. Allergy Appl Immunol 1974;47:623–632.
368. Pineda AA, Taswell HF, Brzica SM, Jr: Delayed Hemolytic 392. Yachnin S, Ruthenberg JM: The initiation and enhancement of
Transfusion reaction. An immunologic hazard of blood trans- human red cell lysis by activators of the first component of
fusion. Transfusion 1978;18:1–7. complement and by first component esterase; studies using
369. Redman M, Regan F, Contreras M: A prospective study of the normal red cells and red cells from patients with paroxysmal
incidence of red cell alloimmunisation following transfusion. nocturnal hemoglobinuria. J Clin Invest 1965;44:518–534.
Vox Sang 1996;71:216–220. 393. Yachnin S: The hemolysis of red cells from patients with
370. Constantoulakis M, Kay HEM: Observations on the centrifu- paroxysmal nocturnal hemoglobinuria by partially purified
gal segregation of young erythrocytes: A possible method of subcomponents of the third complement component. J Clin
genotying the transfused patient. J Clin Path 1963;12:312. Invest 1965;44:1534–1546.
371. Renton PH, Hancock JA: A simple method of separating ery- 394. Ness PM, Shirey RS, Thoman SK, Buck SA: The differentiation
throcytes of different ages. Vox Sang 1964;9:183. of delayed serologic and delayed hemolytic transfusion reac-
Unusual Aspects of Acquired Immune Hemolytic Anemias 373

tions: Incidence, long-term serologic findings, and clinical 418. Kickler TS, Ness PM, Herman JH, Bell WR: Studies on the patho-
significance. Transfusion 1990;30:688–693. physiology of posttransfusion purpura. Blood 1986;68:347–350.
395. Dameshek W, Levine P: Isoimmunization with Rh factor in 419. Dieleman LA, Brand A, Claas FH, van de KC, Witvliet M,
acquired hemolytic anemia. N Engl J Med 1943;228:641–644. Giphart MJ: Acquired Zwa antigen on Zwa negative platelets
396. Allen FH: Proceedings of the 8th Congress of the International demonstrated by western blotting. Br J Haematol 1989;
Society of Blood Transfusion, 1960:359. 72:539–542.
397. Weiner AS: Hemolytic reactions following transfusions of 420. McCrae KR, Herman JH: Posttransfusion purpura: Two unusual
blood of the homologous group. Arch Pathol 1941;32:227–250. cases and a literature review. Am J Hematol 1996;52:205–211.
398. Gajewski JL, Petz LD, Calhoun L, et al: Hemolysis of transfused 421. Bussel JB, Zabusky MR, Berkowitz RL, McFarland JG: Fetal
group O red blood cells in minor ABO-incompatible unrelated- alloimmune thrombocytopenia. N Engl J Med 1997;337:22–26.
donor bone marrow transplants in patients receiving 422. Beutler E: Paroxysmal nocturnal hemoglobinuria. In Beutler E,
cyclosporine without posttransplant methotrexate. Blood Lichtman MA, Coller BS, Kipps TJ, Seligsohn U (eds): Williams’
1992;79:3076–3085. Hematology, 6th ed. New York: McGraw-Hill, 2001:419–424.
399. Greeno EW, Perry EH, Ilstrup SJ, Weisdorf DJ: Exchange 423. Rosse WF: Paroxysmal nocturnal hemoglobinuria: The bio-
transfusion the hard way: Massive hemolysis following trans- chemical defects and the clinical syndrome [review]. Blood
plantation of bone marrow with minor ABO incompatibility. Rev 1989;3:192–200.
Transfusion 1996;36:71–74. 424. Nakakuma H: Mechanism of intravascular hemolysis in
400. Herzog P, Korinkova P, Stambergova M, Lukasova M: Auto paroxysmal nocturnal hemoglobinuria (PNH). Am J Hematol
anti-A1 and auto anti-NA1 after bone marrow transplanta- 1996;53:22–29.
tion. Folia Haematol Int Mag Klin Morphol Blutforsch 425. Nakakuma H, Hidaka M, Nagakura S, et al: Expression of
1987;114:874–880. cryptantigen Th on paroxysmal nocturnal hemoglobinuria
401. Toren A, Dacosta Y, Manny N, et al: Passenger B-lymphocyte- erythrocytes in association with a hemolytic exacerbation. J
induced severe hemolytic disease after allogeneic peripheral Clin Invest 1995;96:201–206.
blood stem cell transplantation. Blood 1996;87:843–844. 426. Sirchia G, Ferrone S, Mercuriali F: Leukocyte antigen-
402. Oziel-Taieb S, Faucher-Barbey C, Chabannon C, et al: Early antibody reaction and lysis of paroxysmal nocturnal hemo-
and fatal immune haemolysis after so-called “minor” ABO- globinuria erythrocytes. Blood 1970;36:334–336.
incompatible peripheral blood stem cell allotransplantation. 427. Dacie JV: Clinicopathological Conference: A fatal case of
Bone Marrow Transplant 1997;19:1155–1156. paroxysmal haemoglobinuria demonstrated at the Post-
403. Bolan CD, Childs RW, Procter JL, Barrett AJ, Leitman SF: graduate Medical School of London. Br Med J 1959;2:559.
Massive immune haemolysis after allogeneic peripheral blood 428. Crosby WH, Stefanini M: Pathogenesis of the plasma transfu-
stem cell transplantation with minor ABO incompatibility. Br sion reaction with especial reference to the blood coagulation
J Haematol 2001;112:787–795. system. J Lab Clin Med 1952;40:374–386.
404. Tiplady CW, Fitzgerald JM, Jackson GH, Conn JS, Proctor SJ: 429. Dameshek W, Neber J: Transfusion reactions to a plasma con-
Massive haemolysis in a group A recipient of a group O stituent of whole blood: Their pathogenesis and treatment by
peripheral blood stem cell allogeneic transplant. Transfus washed red blood cell transfusions. Blood 1950;5:129–147.
Med 2001;11:455–458. 430. Crosby WH, Dameshek W: Paroxysmal nocturnal hemoglo-
405. Worel N, Greinix HT, Keil F, et al: Severe immune hemolysis binuria: The mechanism of hemolysis and its relation to the
after minor ABO-incompatible allogeneic peripheral blood coagulation mechanism. Blood 1950;5:822–842.
stem cell transplantation occurs more frequently after non- 431. Dacie JV, Firth D: Blood transfusion in nocturnal haemoglo-
myeloablative than myeloablative conditioning. Transfusion binuria. Br Med J 1943;1:626–628.
2002;42:1293–1301. 432. Heffernan CK, Jaswon N: A case of paroxysmal nocturnal
406. Petz LD, Calhoun L, Shulman IA, Johnson C, Herron RM: The haemoglobinuria associated with secondary haemochromato-
sickle cell hemolytic transfusion reaction syndrome. Trans- sis, a lower nephron nephrosis, and a megaloblastic anaemia.
fusion 1997;37:382–392. J Clin Pathol 1955;8:211–217.
407. King KE, Shirey RS, Lankiewicz MW, Young-Ramsaran J, 433. Heitzman EJ, Campbell JS, Stefanini M: Paroxysmal nocturnal
Ness PM: Delayed hemolytic transfusion reactions in sickle hemoglobinuria with hemosiderin nephrosis. Am J Clin Path
cell disease: Simultaneous destruction of recipients’ red cells. 1953;23:975–986.
Transfusion 1997;37:376–381. 434. Baranett EC, Dunlop JB, Pullar TH: Chronic haemolytic
408. Reed W, Walker P, Haddix T, Perkins HA: Acute anemic anaemia with paroxysmal haemoglobinuria (Marchiafava
events in sickle cell disease. Transfusion 2000;40:267–273. syndrome). Report of a case improved by splenectomy. N Z
409. Garratty G: Severe reactions associated with transfusion of Med J 1951;50:39–43.
patients with sickle cell disease. Transfusion 1997;37:357–361. 435. Hirsch J, Ungar B, Robinson JS: Paroxysmal nocturnal haemo-
410. Win N, Doughty H, Telfer P, Wild BJ, Pearson TC: globinuria: An acquired dyshaemopoiesis. Austral Ann Med
Hyperhemolytic transfusion reaction in sickle cell disease. 1964;13:24–31.
Transfusion 2001;41:323–328. 436. Dacie JV: Transfusion of saline-washed red cells in nocturnal
411. Sirchia G, Morelati F, Rebulla P: The sickle cell hemolytic haemoglobinuria (Machiafava-Micheli disease). Clin Sci
transfusion reaction syndrome [letter]. Transfusion 1997; 1948;7:65–75.
37:1098–1099. 437. Sirchia S, Zanella A: Transfusion of PNH patients. Transfusion
412. Greene D, Khan S: A phenomenon of delayed hemolytic trans- 1990;30:479.
fusion reaction [abstract]. Transfusion 1991;31:23S. 1991. 438. Rosse WF: Paroxysmal nocturnal hemoglobinuria as a molec-
413. Shulman NR: Posttransfusion purpura: Clinical features and the ular disease. Medicine (Baltimore) 1997;76:63–93.
mechanism of platelet destruction. In Nance ST (ed): Clinical 439. Brecher ME, Taswell HF: Paroxysmal nocturnal hemoglobin-
and Basic Science Aspects of Immunohematology. Arlington, uria and the transfusion of washed red cells. A myth revisited.
VA: American Association of Blood Banks, 1991:137–154. Transfusion 1989;29:681–685.
414. Mueller-Eckhardt C: Post-transfusion purpura. Br J Haematol 440. Salama A, Mueller-Eckhardt C: Binding of fluid phase C3b to
1986;64:419–424. nonsensitized bystander human red cells: A model for in vivo
415. Lau P, Sholtis CM, Aster RH: Post-transfusion purpura: An effects of complement activation on blood cells. Transfusion
enigma of alloimmunization. Am J Hematol 1980;9:331–336. 1985;25:528–534.
416. George JN, Rizvi MA: Thrombocytopenia. In Beutler E, 441. Gocke DJ: In vitro studies of the plasma requirement in
Lichtman MA, Coller BS, Kipps TJ, Seligsohn U (eds): Williams’ platelet damage by an unrelated antigen-antibody reaction.
Hematology, 6th ed. New York: McGraw-Hill, 2001:1495–1539. Fed Proc 1964;23:404.
417. George JN, Pickett EB, Heinz R: Platelet membrane micropar- 442. Test ST, Woolworth VS: Defective regulation of complement
ticles in blood bank fresh frozen plasma and cryoprecipitate. by the sickle erythrocyte: Evidence for a defect in control of
Blood 1986;68:307–309. membrane attack complex formation. Blood 1994;83:842–852.
374 Immune Hemolytic Anemias

443. Crookston MC, Tilley CA: A and B Lewis antigens in plasma. 455. Ohto H, Yasuda H, Abe R, et al: Platelet transfusion refrac-
Proceedings of the Fifth International Convocation on toriness associated with plasma proteins [letter comment].
Immunologics. Basel Karger, 1977:246–256. Brit J Haemat 1993;83:680–681.
444. Hostrup H: A and B blood group substances in the serum of 456. Heal JM, Cowles J, Masel D, Rowe JM, Blumberg N:
normal subjects. Vox Sang 1962;7:701–721. Antibodies to plasma proteins: an association with platelet
445. Holburn AM, Masters CA: The radioimmunoassay of serum transfusion refractoriness. Br J Haematol 1992;80:83–90.
and salivary blood group A and Lea glycoproteins. Brit J 457. Castellino SM, Combs MR, Zimmerman SA, Issitt PD, Ware
Haemat 1974;28:157–167. RE: Erythrocyte autoantibodies in paediatric patients with
446. Hartmann G: Blood Group Antigens in Human Organs. sickle cell disease receiving transfusion therapy: Frequency,
Copenhagen, Munksgaard, 1941. characteristics and significance. Br J Haematol 1999;104:
447. Renton PH, Hancock JA: Uptake of A and B antigens by trans- 189–194.
fused group O erythrocytes. Vox Sang 1962;7:33. 458. Marks JD, Ouwehand WH, Bye JM, et al: Human antibody
448. Race RR, Sanger R: Blood Groups in Man, 6th ed. New York: fragments specific for human blood group antigens from a
Blackwell Scientific, 1975. phage display library. Biotechnology (NY) 1993;11:1145–1149.
449. Bird GWG, Battey DA, Greenwell P, et al: Further observations 459. Ouwehand WH, Bye JM, Gorick BD, et al: The humoral
of the Birmingham chimaera. J Med Genet 1976;13:70–71. immune response against blood group antigens at the molec-
450. Szymanski IO, Tilley CA, Crookston MC, Greenwalt TJ, ular level. Vox Sang 1994;67(Suppl 3):7–12.
Moore S: A further example of human blood group chi- 460. Thompson KM, Sutherland J, Barden G, et al: Human mono-
maerism. J Med Genet 1977;14:279–281. clonal antibodies specific for blood group antigens demon-
451. Maeda K, Taniwaki K, Santo T, et al: Anti-A and/or anti-B is not strate multispecific properties characteristic of natural
detectable in some patients who underwent ABO-incompatible autoantibodies. Immunology 1992;76:146–157.
bone marrow transplantation. Transfusion 1995;35:635–639. 461. Barker RN, Elson CJ: Multiple self epitopes on the Rhesus
452. Arndt PA, Leger RM, Garratty G, Calhoun L, Chow Y, Petz polypeptides stimulate immunologically ignorant human T
LD: Use of flow cytometry for detecting uptake of A/B blood cells in vitro. Eur J Immunol 1994;24:1578–1582.
group substance on transfused/transplanted O RBCs 462. Shirey RS, Ness PM: New concepts of delayed hemolytic
[abstract]. Transfusion 199;39(Suppl):43S–44S. transfusion reactions. In Nance SJ (ed): Clinical and Basic
453. Safai-Kutti S, Zaroulis CG, Day NK, Good RA, Kutti J: Platelet Science: Aspects of Immunohematology. Arlington, VA:
transfusion therapy and circulating immune complexes. Vox American Association of Blood Banks, 1991:179.
Sang 1980;39:22–27. 463. Issitt PD: Applied Blood Group Serology. Miami:
454. Kutti J, Zaroulis CG, Safai-Kutti S, Dinsmore RE, Day NK, Montgomery Scientific, 1985.
Good RA: Evidence that circulating immune complexes 464. Morrison FS, Mollison PL: Post-transfusion purpura. N Engl J
remove transfused platelets from the circulation. Am J Med 1966;275:243–248.
Hematol 1981;11:255–259.
C H A P T E R 1 0

Blood Transfusion in
Autoimmune Hemolytic
Anemias

Patients with autoimmune patients with AIHA. We next review the methods
hemolytic anemia (AIHA) that should be employed for the optimal selection of
often present with anemia of blood for patients with autoantibodies. Finally, we
sufficient severity to suggest a discuss the optimal volume of blood to be transfused
need for blood transfusion. (which can be of critical importance in safely treating
When anemia of such severity patients with severe hemolytic anemia), in vivo
is discovered, physicians fre- compatibility testing, the use of warm blood for
quently refer a sample of patients with cold antibody AIHAs, the use of
blood to the blood transfusion RBC substitutes, the use of washed or leukocyte-
laboratory while simultane- reduced RBCs, and the use of autologous blood for
ously initiating diagnostic studies to determine the transfusion.
cause of the anemia. Indeed, a diagnosis of AIHA is
often first made by the blood transfusion service when
autoantibodies are detected during the performance ASSESSING THE NEED FOR
of the compatibility test. TRANSFUSION IN PATIENTS WITH
Transfusion of patients with AIHA presents a unique AUTOIMMUNE HEMOLYTIC ANEMIA
set of potential problems. The indications for transfu-
sion must be considered in light of the following facts:
As with all clinical decisions concerning therapy, the
• The risks of transfusion are somewhat increased possible benefits must be weighed in relationship to
due to the difficulty of compatibility testing. potential risks. The advisability of blood transfusion is
• The patient’s autoantibody can be expected to related to the severity of the anemia, to the determi-
cause a shortened life span of transfused red blood nation of whether the anemia is rapidly progressive,
cells (RBCs). and especially to the associated clinical findings.
Although, as indicated previously, the risks associated
Nevertheless, blood should never be denied to a with transfusion for patients with AIHA are greater
patient with a justifiable need, even though the com- than for patients without immunohematologic abnor-
patibility test might be strongly positive.1-5 malities, the risk of not transfusing is even greater for
In this chapter, we first discuss the principles most patients with severe anemia, and transfusion is
concerning indications for blood transfusions for mandatory in patients with life-threatening anemia.
375
376 Immune Hemolytic Anemias

A major difference between transfusing patients to transfuse even those with severe anemia. Examples
with AIHA and those without RBC autoantibodies is of such cases have been reported by Conley and asso-
that the clinician must consider the time that is ciates,8,9 who described five patients with AIHA and
required by the transfusion service to do complex reticulocytopenia who developed life-threatening
serologic tests to assure that the optimal RBC product anemia but were not transfused because physicians
is obtained. The clinician and the transfusion service were concerned that compatible blood could not be
must be in contact with one another so that the trans- obtained. This was true even though the patients’
fusion service understands the urgency of the situa- hematocrits were at levels of 8%–10%. After transfer
tion and the clinician understands the complexity of to a tertiary care medical center, the patients were
the serologic studies to be undertaken. In a large promptly transfused—a measure that the authors felt
majority of cases, adequate time can be taken to com- was unquestionably life saving.
plete appropriate antibody identification studies and Patients with AIHA and reticulocytopenia are
compatibility tests. In extremely urgent situations, it in a particularly precarious position. Crosby and
could be necessary to take added risks, but even here, Rappaport10 reported on 15 such patients who were
cautionary measures can minimize these risks. not transfused; 12 of the 15 patients died. Numerous
Nowhere in the management of patients with other authors have described reticulocytopenia in
immune hemolytic anemias is the communication patients with AIHA.11-15 In some instances, the reticu-
between clinician and laboratory personnel more locytopenia is caused by human parvovirus B19.16-18
important than with regard to blood transfusion. In other cases, it could have an immune basis, and in
Because the serologic evaluation potentially can be many instances there is no identifiable cause19,20 (see
time consuming, notification of the potential need for also Chapter 3).
transfusion and initiation of work-up should occur as
soon as transfusion is considered.6,7 Results of evalua- Assessing the Acuteness of Onset
tions performed at other facilities (e.g., prior identi-
fication of alloantibodies or determination of the and Rapidity of Progression of AIHA
patient’s RBC phenotype) can expedite the prepara- When a patient presents with AIHA and a moderately
tion of blood. The knowledge of recent transfusions is severe anemia, it is not possible to predict with cer-
also important for the transfusion service laboratory tainty whether the anemia will rapidly become more
to determine the optimal serologic procedures for severe. Thus, serial determinations of the hemoglobin
identifying RBC alloantibodies in the presence of and hematocrit should be performed at intervals
broadly reactive autoantibodies. determined by the evaluation of the severity of the
Other important information should include a illness. In particular, the physician should note
history of pregnancy and of prior transfusions. whether the patient appears acutely ill with symp-
Clinicians frequently forget that the presence of RBC toms attributable to acute hemolysis—fever, malaise,
alloantibodies is extremely unlikely if the patient has and pain in the back, abdomen, and legs.21 It is also
never been pregnant or previously received a transfu- important to note whether hemoglobinuria and
sion. Transfusion medicine specialists are reluctant to hemoglobinemia are present. These findings are
depend on such information because the medical usually manifestations of severe hemolysis.
history might not be reliable. Indeed, hospitalized If a patient is acutely ill or has a history of an abrupt
patients may have been transfused without having onset of the illness, or if visual inspection of serum and
been aware of the transfusion. Nevertheless, with urine indicate hemoglobinemia and/or hemoglobin-
some patients one can be quite sure that there have uria, the hematocrit level should be determined every 2
been no previous transfusions or pregnancies, and to 4 hours initially. In less acutely ill patients, initial
this information reassures the physician that no testing may take place at 12- to 24-hour intervals. The
alloantibody-induced hemolytic transfusion reaction frequency of testing may soon be decreased if the sever-
will occur. ity of the anemia proves to be essentially constant. In
Finally, it must be emphasized that the decision to some cases of fulminant hemolysis, a significant fall in
transfuse should depend not on the serologic findings the hematocrit can occur within hours, whereas in a
but rather on an evaluation of the patient’s clinical majority of patients with AIHA, the anemia is more
status. The patient’s physician should assess the slowly progressive.
acuteness of onset and the rapidity of progression of
the anemia, the patient’s symptoms and signs caused
by the anemia, and the probable effectiveness of THE APPROPRIATE USE OF BLOOD
therapy other than transfusion (see Chapter 11). IN VARIOUS CLINICAL SETTINGS
IN PATIENTS WITH AIHA
Reluctance to Transfuse
Patients with AIHA Stable Anemia during Initial Evaluation
Perhaps one of the most common mistakes in the Patients with an anemia that is essentially stable
management of patients with AIHA is the reluctance during the initial period of evaluation often need not
Blood Transfusion in Autoimmune Hemolytic Anemias 377

be transfused, even though they might have symp- Chronic Stable Anemia
toms such as a decrease in exercise tolerance or palpi-
tations with exertion. Furthermore, the response to Many patients (especially those with cold agglutinin
therapy or spontaneous improvement might be rapid. syndrome) are able to compensate partially for their
For example, 50% of the patients with warm-antibody shortened RBC survival and maintain a relatively
autoimmune hemolytic anemia (WAIHA) will respond stable (albeit occasionally quite severe) degree of
to adequate doses of corticosteriods during the first anemia. For such patients, for whom transfusion may
week of therapy (Chapter 11), and acute paroxysmal be considered as a means of relieving symptoms, the
cold hemoglobinuria (PCH) seldom lasts longer than advantages and disadvantages of such management
7 to 10 days.22 must be weighed carefully. The hazards and incon-
venience of long-term transfusions must be consi-
dered, and it is often preferable for the patient to live
Progressively Severe Anemia with some symptoms of anemia rather than to face the
Some patients have an anemia that is steadily pro- hazards of long-term transfusion therapy.23
gressive in severity, leading to the development of In some patients, in spite of adequate therapy,
symptoms of hypoxemia. In association with an eva- hemolysis proceeds chronically at a rate greater than
luation of the clinical status of the patient, laboratory that of their RBC production. In this situation, chronic
values offer some guidance as to the necessity for transfusion is necessary to sustain life, and the atten-
transfusion (Table 10-1). If the hemoglobin level is dant risks, cost, and inconvenience must be accepted.
above 10 g/dL, transfusion therapy is almost never Transfusions will at least partially suppress erythro-
indicated. Even at a level of 8–10 g/dL, transfusion is poiesis, and the frequency of transfusions will need to
rarely necessary or desirable. Clinical judgment is be determined arbitrarily and will depend on the rate
most critical at a hemoglobin level of 6–8 g/dL. At a of hemolysis. It is usually convenient to give two to
level of hemoglobin level below 6 g/dL, most patients three units of packed RBCs at a time when necessary.
require transfusion. It is not advisable to correct the anemia completely,
Manifestations of life-threatening anemia can occur, and transfusion to a level of hemoglobin of 8–11 g/dL
such as progressively severe angina, cardiac decom- is perhaps optimal.23
pensation, or neurologic symptoms that include
marked lethargy, weakness, somnolence, and mental
confusion. These symptoms usually occur when the
hemoglobin level is below 5 g/dL but can develop Fulminant Hemolytic Anemia
even with less severe anemia. The presence of such Least common among the indications for transfusion
symptoms indicates an urgent need for transfusion, in AIHA is rapidly progressive anemia caused by
but transfusion should be not be withheld until those acute massive hemolysis. Nevertheless, such fulmi-
symptoms arise. For patients with anemia of such nant hemolysis does occur, and such patients might
severity, or for those patients whose rate of progres- even be hypotensive.
sion of anemia indicates that this level of severity will Patients who have AIHA of such severity are likely
probably be reached, RBCs are needed urgently. to have gross evidences of hemolysis, particularly
Indeed, death could result from progressive anemia. hemoglobinuria and hemoglobinemia. Diagnoses
In the management of these acutely ill patients, the such as PCH, Clostridium perfringens (welchii) sep-
transfusion of RBCs sufficient to maintain a modest ticemia, and drug-induced immune hemolytic
increase in hematocrit until therapy for the AIHA anemia should be investigated quickly. The onset can
becomes effective is probably optimal. be so acute that a reticulocytosis might not be
present, as the bone marrow might not have had
time to compensate. Indeed, an increase in the retic-
TABLE 10-1. GUIDELINES FOR ASSESSING PHYSIO- ulocyte count in response to a sudden decrease in
LOGICAL IMPAIRMENT OF THE ANEMIC PATIENT RBC mass can require 7 to 10 days.24
AND PRESCRIBING TRANSFUSION STRATEGY Although such patients are uncommon, transfusion
for them is urgent. If manifestations of shock are
Average Probability of present, the immediate aim of transfusion is to
Hemoglobin Significant
Level (g/dL) Impairment Transfusion Strategy improve vital signs, which can be restored temporar-
ily by the use of electrolyte or colloid solutions.
≥10 Very low Avoid Simultaneously, the physician should communicate a
8–10 Low Avoid; transfuse only if demonstrably sense of urgency to the blood transfusion laboratory
better after transfusion trial
6–8 Moderate Try to avoid by decreased activity;
to find RBCs for transfusion. Rarely will these RBCs
if impossible, transfuse be “compatible,” but, nevertheless, transfusion is
≤6 High Frequently requires transfusion mandatory in this acute, life-threatening context. A
possible alternative to RBC transfusion is the use of
From Petz LD: Blood transfusion in acquired hemolytic anemia. In Petz LD,
Swisher SN, Kleinman S, Spence RK, Strauss RG (eds): Clinical Practice of effective oxygen-carrying solutions, should they
Transfusion Medicine, 3rd ed. New York: Churchill Livingstone, 1996:469–499. become available and practical (also see p. 395).25,26
378 Immune Hemolytic Anemias

THE RISKS OF TRANSFUSION FOR sion of patients with AIHA (see the section on the
PATIENTS WITH AUTOIMMUNE optimal volume of blood to be transfused).3,34,37
HEMOLYTIC ANEMIA
COMPATIBILITY TESTING
Risks Caused by the Patient’s IN WARM-ANTIBODY AIHA
Autoantibody
The selection of blood for transfusion to patients with
For patients with AIHA, the risks of blood transfusion WAIHA is one of the most difficult tasks faced by a
beyond the usual risks relate to the presence of the blood transfusion laboratory. A significant commit-
patient’s RBC autoantibody. Autoantibodies usually ment of time can be required to resolve the problems
react with all normal RBCs, with the result that any presented by a patient with broadly reactive autoanti-
transfused RBCs are likely to have a shorter-than- bodies. We have divided the discussion of these prob-
normal life span. This reaction cannot be avoided, lems into a consideration of RBC phenotyping, the
assuming that optimal therapeutic measures are being detection of alloantibodies, and the significance of
used to treat the AIHA. The RBC autoantibody might autoantibody specificity.
react strongly in vitro (e.g., 2+ to 4+ by indirect
antiglobulin test [IAT]) with all available donor RBCs
to be transfused, thus making it impossible to obtain Red Cell Phenotyping and Genotyping
compatible blood for transfusion. Nevertheless, acute Techniques for ABO and Rh cell typing and extended
symptomatic transfusion reactions occur only infre- phenotyping in patients with AIHA are discussed in
quently.27,28 Survival of transfused RBCs is about as Chapter 6. For all patients with WAIHA who require
good as survival of the patient’s own RBCs, and the transfusion, we strongly recommend determining the
net result is that transfusion generally causes tempo- extended RBC phenotype of the patient prior to the
rary benefit. initial transfusion. If time does not allow, it is never-
Although this finding is generally true, some theless advisable to obtain a blood specimen from the
patients appear to derive no benefit from transfusion29 patient for subsequent typing. This information is par-
or, in very unusual instances, they develop significant ticularly useful in the management of AIHA.7 One
complications such as acute renal failure,30,31 dissemi- must keep in mind that determining the patient’s RBC
nated intravascular coagulation, or post-transfusion phenotype can be impossible after the patient has
hemoglobinuria.33-36 Such complications are probably received a transfusion. Therefore, one should not miss
most likely to occur if relatively large volumes of the opportunity when the patient is first examined.
blood are given and if the patient has brisk hemolysis. Knowledge of the patient’s extended RBC phenotype
provides valuable information when techniques to
Risks Caused by Alloantibodies identify serum alloantibodies cannot be performed or
produce inconclusive results. Blood for transfusion can
When the patient’s serum reacts with all RBCs in potentially be selected that lacks those antigens against
routine compatibility and antibody identification which the patient could have produced a clinically
tests, the blood transfusion laboratory must use addi- significant antibody. For example, if the patient’s Rh
tional techniques in an attempt to demonstrate RBC phenotype is known, it is usually feasible to select
antibodies other than the autoantibody. This is a criti- donor units of the same phenotype to avoid alloanti-
cal aspect of selection of donor blood because, if the body-induced hemolysis. Also, if the patient is, for
patient has previously been transfused or has been example, Jk(a+), one need not be concerned about anti-
pregnant and has developed RBC alloantibodies (e.g., Jka alloantibodies; if the patient is K negative, the use of
anti-D, anti-K, anti-Jka), donor RBCs lacking such K-negative blood would eliminate concern about the
antigens must be selected for transfusion to prevent a possible presence of anti-K4 (also see the section later in
severe alloantibody-induced hemolytic transfusion this chapter on the use of phenotpyically matched RBC
reaction. Such compatibility testing procedures are at for transfusion). Also, knowledge of the patient’s RBC
times complex and can be difficult to resolve phenotype can make selection of RBCs for allogeneic
definitively. adsorption studies significantly easier (see the discus-
sion later in this chapter).
Risks Caused by the Increase in RBC Unfortunately, phenotyping for all RBC antigens can
be difficult when the direct antiglobulin test (DAT) is
Mass as a Result of Transfusion positive. The use of monoclonal typing reagents is of
Post-transfusion hemoglobinemia and hemoglobinuria significant assistance, as are special techniques with the
have generally been attributed to an increased rate of intent of removing the autoantibody without altering
hemolysis, although they could more commonly antigen expression (see Chapter 6).38
occur as a result of the increase in the total mass of In the case of previously transfused patients, one
RBCs available for destruction.33-35 Awareness of this may separate reticulocytes from the other RBCs and
risk has led to strong warnings against overtransfu- perform typing on them to determine the patient’s phe-
Blood Transfusion in Autoimmune Hemolytic Anemias 379

notype. Branch and colleagues39 described a rapid tech-


nique for the age-fractionation of human RBCs into TABLE 10-2. RBC ALLOANTIBODIES IN PATIENTS
reticulocyte-enriched (young) RBCs and reticulocyte- WITH WARM AUTOANTIBODIES
poor (old) RBCs. Using this method, it was possible to
Number of Percentage
perform accurate RBC typing for six patients within Alloantibodies Found/ of Sera with
12 hours after transfusion of 4 to 29 units of blood Reference Number of Sera Tested Alloantibodies
within a 12-hour period. Other methods for reticulo-
cyte separation using microhematocrit centrifugation Morel et al47 8/20 40
have also been described.40-42 In patients with a positive Branch and Petz48 5/14 36
Wallhermfechtel et al49* 19/125 16
DAT, one might also need to first remove IgG from the Laine and Beattie50* 41/109 38
surface of the RBC before typing. These complexities James et al51* 13/41 32
should inspire one to perform phenotyping prior to the Issitt et al (alloadsorptions)52† 13/34 38
first transfusion. Issitt et al (autoadsorptions)52† 5/41 12
Leger and Garratty53‡ 105/263 40
More recently, methods for genotyping RBCs using
DNA typing have become available.43-45 Castilho and Totals 209/647 32
coworkers45 tested RBCs from 15 patients with * Forty-two to 77 percent of alloantibodies were not evident before adsorption.
WAIHA, all of whom had been phenotyped by † Calculations excluded autoantibodies that mimicked alloantibodies.
‡ Sera were first tested for autoantibodies using low-lonic-strength saline
hemagglutination for Rh, Kell, Kidd, and Duffy after solution. When autoantibodies were initially detected using PEG, 47 percent
antibody dissociation with chloroquine diphosphate. of sera contained alloantibodies.
The results for all 15 patients using hemagglutination From Branch DR, Petz LD: Detecting alloantibodies in patients with
autoantibodies. Transfusion 1999;39:6–10.
were unclear, probably due to insufficient dissociation
of antibodies by the chloroquine treatment. In con-
trast, genotyping results were clear, even for patients
who had been transfused because the large excess of clinically significant alloantibodies in 10% of sera.
patients’ DNA prevented detection of DNA from This study is impossible to compare with other
transfused leukocytes. reports because it included sera with both warm-
and cold-reactive autoantibodies. Also, the authors
pointed out that their policy is to issue K-negative
Detection of Alloantibodies blood of matched Rh phenotype, and they speculate
If the patient’s autoantibody reacts with all normal that this could have contributed to the relatively low
RBCs, several available techniques allow one to detect occurrence of alloantibodies in their series. For these
alloantibodies that might also be present. These pro- reasons, their data were not included in Table 10-2.
cedures are quite practical and should be performed So and associates54 reported a rate of alloimmuniza-
in all cases except when extreme emergency precludes tion of 11.3% in a Chinese population. They suggested
adequate evaluation before transfusion. that the comparatively low rate of alloimmunization
probably reflects the genetic homogeneity among
Chinese people with respect to RBC phenotypes.
The Incidence of Alloantibodies
in Patients with AIHA Who
Require Transfusion Methods for Detection of RBC
In seven reported studies of randomly selected sera
Alloantibodies in Patients with
containing warm autoantibodies, investigators have Autoantibodies
found alloantibodies of potential clinical significance COMPARISON OF DIRECT AND INDIRECT
in 12%–40% of samples (Table 10-2).46-53 In some ANTIGLOBULIN TESTS
instances, the alloantibodies are evident even in the
presence of broadly reacting autoantibodies because A comparison of the DAT and IAT sometimes affords
of variable reactivity when tested against an RBC extremely valuable information. In patients with
panel. Three reports, however, indicated that, in WAIHA, the IAT caused by the autoantibody is gen-
42%–77% of sera, alloantibodies were not evident erally weaker than the DAT. Apparently, most of the
before adsorption.49-51 Because alloantibodies were autoantibody is adsorbed to the patient’s RBCs in
detected in 209 of 647 sera (32%) (see Table 10-2), there vivo. A coexisting alloantibody will not, of course, be
is an obvious need for a method to detect these antibo- adsorbed by the patient’s RBCs and might result in a
dies to prevent alloantibody-induced hemolytic trans- strongly positive IAT. Thus, if the IAT is significantly
fusion reactions. Indeed, undetected alloantibodies stronger than the DAT, the presence of an alloanti-
could be the cause of increased hemolysis following body in association with the autoantibody is strongly
transfusion, which might falsely be attributed to an suspected.
increase in the severity of AIHA.4 If the DAT is equally strong or stronger than the
Two reports were not included in the foregoing IAT, no conclusion can be reached concerning the
summary. Sokol and colleagues28 reported finding presence or absence of alloantibodies.
380 Immune Hemolytic Anemias

TESTING PATIENT’S SERUM AGAINST ADSORPTION PROCEDURES—OVERVIEW


A RED CELL PANEL
Adsorption with autologous RBCs (”warm autoadsorp-
If the screening tests for serum antibody reveal an tion”) is the optimal method for the detection of all
antibody reactive at 37°C, the serum should be tested potentially significant alloantibodies, as it uses the
against a panel of phenotyped RBCs, as is routine in patient’s RBCs to adsorb the autoantibody from the
any determination of alloantibody specificity. If a serum, leaving only the alloantibodies. This is parti-
weakly reactive autoantibody and a strongly reactive cularly important when clinically significant alloanti-
alloantibody are present, the differences in the bodies to high-frequency antigens are present. A
strength of the reaction of various cells of the panel further advantage of adsorption with autologous
will make this evident. For example, Table 10-3 shows RBCs compared with allogeneic adsorptions is that
a strong anti-K together with a weak autoantibody. adsorption with only one sample of RBCs is needed.
One has no assurance, however, that a patient’s This results in a greater volume of serum remaining
alloantibody will react more strongly than the autoan- after adsorption than is the case after adsorption with
tibody, so additional tests to detect alloantibody are multiple cells, as is generally necessary with allo-
necessary. geneic adsorptions. Indeed, unless one begins with a
generous supply of serum, it can be difficult to
perform specificity tests and cross-match tests with
the small volume of serum remaining after adsorption
DILUTION TECHNIQUE tests with multiple samples of allogeneic RBCs. These
advantages are balanced by the fact that it might be
One may select a dilution of the patient’s serum that difficult to obtain a sufficiently large sample of RBCs
reacts approximately 1+ in the IAT, and then test that for autoadsorption studies from severely anemic
dilution against a panel of RBCs. Table 10-4 illus- patients. This is a particular problem for reference
trates the results obtained in a case in which the laboratories, which, therefore, often must rely on the
alloantibody of anti-c specificity had a titer of 64 and allogeneic adsorption procedure.
the autoantibody a titer of only 16. When the undi- It is of no value to incubate patient’s serum and RBCs
luted serum was tested, all cells reacted 3+, but a 1:64 in vitro to adsorb autoantibody, as this has occurred in
dilution of the patient’s serum reacted only with c+ vivo. It is assumed that autoantibody is present in the
RBCs. Although this technique frequently provides serum because all autoantigen sites on the patient’s
useful information, it is informative only when the RBCs are saturated. For effective autoadsorption, some
alloantibody is of higher titer than the autoantibody, of the antibody coating the patient’s RBCs should be
and therefore one cannot rely on it. removed before the procedure. To accomplish this, a
Oyen and Angeles55 recommended that sera with heat-elution technique was used originally; the patient’s
autoantibodies be diluted 1-in-5 to screen for under- heat-eluted RBCs were then enzyme-treated to enhance
lying alloantibodies. The success of this approach adsorption of autoantibody.47,56 Currently, the most
requires not only that the autoantibody be in lower widely used procedure uses “ZZAP” reagent.48 ZZAP
titer than the alloantibody but also that the autoanti- consists of a mixture of 0.1 M dithiothreitol (DTT) plus
body be essentially nonreactive at the suggested 0.1% cysteine-activated papain or 0.1% ficin; it effec-
dilution. Leger and Garratty53 evaluated the effec- tively dissociates IgG from RBCs and simultaneously
tiveness of this method and reported that only 5 of 26 results in their treatment with a proteolytic enzyme.
(19%) potentially clinically significant alloantibodies A less labor-intensive option would be autoadsorp-
were identifiable (Table 10-5). Fourteen potentially tion without prior treatment of the patient’s RBCs;
clinically significant alloantibodies (54%) were not this could be feasible using polyethylene glycol
identified because they continued to be masked by (PEG).53,57,58 Although Liew and Duncan57,58 did find
the autoantibody after dilution. In addition, 7 of the the method effective in their study of six patients, the
26 alloantibodies (27%) were not detected because adsorbing RBCs that they used were also pretreated
completely negative results were obtained in the with papain.46 Cheng and coworkers59 commented
diluted serum. Alloantibodies missed by the dilution that the method “awaits standardization.” Indeed,
method included Rh system antibodies, anti-Jka, Champagne and Moulds60 reported that an autoad-
-Jkb, -Fya, -s, and -Kn/McC. Also, only 20 of 93 sera sorption using untreated autologous RBCs mixed
(22%) containing warm autoantibodies without with PEG failed to detect an alloanti-K that was
alloantibodies were nonreactive after dilution at a detected when the adsorptions were performed with
ratio of 1:5, indicating that the method is not widely ficin-treated autologous RBCs.
applicable. (Oyen and Angeles55 had reported that 50 Adsorption with allogeneic RBCs (allogeneic adsorp-
of 119 sera [42%] were nonreactive after dilution to 1- tion) is necessary when adsorption studies are to be
in-5.) Thus, it is preferable to select a dilution of the performed on a patient who has had a recent transfu-
patient’s serum that reacts approximately 1+ in the sion, or when an adequate supply of the patient’s RBCs
IAT, rather than using an arbitrarily selected dilution is not available. Allogeneic adsorptions may be per-
such as 1-in-5. formed using ZZAP-treated RBCs48 or by using PEG
TABLE 10-3. REACTIONS OBTAINED WITH A SERUM CONTAINING ALLO ANTI-K AND AN UNDEFINED AUTOANTIBODY
WHEN REACTED WITH A PANEL OF RED CELLS

Donor Rh Sex
No. Phenotype Rh Kell Duffy Kidd Lewis MNS P1 Lutheran Linked Results

C D E c e f Cw V K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb M N S s P1 Lua Lub Xga IC 37 IAT

1 rr 0 0 0 + + + 0 0 0 + 0 + 0 + + 0 + + + 0 + 0 + 0 0 0 + + 0 0 1+
2 rr 0 0 0 + + + 0 0 + 0 0 + 0 + 0 + + 0 0 + + 0 + 0 + 0 + + 0 0 3+
3 rr 0 0 0 + + + 0 0 + + 0 + 0 + + + + 0 + 0 0 + 0 + + + + + 0 0 3+
4 r′r + 0 0 + + + 0 0 0 + 0 + 0 + 0 + + + 0 + 0 + 0 + + 0 + + 0 0 1+
5 r″r 0 0 + + + + 0 0 0 + 0 + 0 + + + 0 + 0 + + + + + + 0 + + 0 0 1+
6 R1wR1 + + 0 0 + 0 + 0 + + 0 + 0 + 0 + 0 + 0 + + 0 0 + 0 0 + + 0 0 3+
7 R1R1 + + 0 0 + 0 0 0 0 + 0 + + + + 0 + 0 0 + + + 0 + + 0 + 0 0 0 1+
8 R0 0 + 0 + + + 0 + 0 + 0 + 0 + + 0 + + 0 0 + + + + + 0 + 0 0 0 1+
9 R2R2 0 + + + 0 0 0 0 0 + 0 + 0 + + 0 + + 0 0 + 0 + + + 0 + 0 0 0 1+
10 R2R2 0 + + + 0 0 0 0 0 + 0 + 0 + 0 + 0 + + 0 0 + + 0 + 0 + 0 0 0 1+

IC = Immediate Centrifugation; 37 = Agglutination at 37°C; IAT = Indirect Antiglobulin Test.


Note that 3+ reactions are obtained with K positive RBCs; all other cells yield 1+ reactions.

TABLE 10-4. RESULTS OF DILUTION TECHNIQUE TO DETERMINE PRESENCE OF ALLOANTIBODY

Dilutions of Patient’s Serum 2 4 8 16 32 64 128 256 512 1024

IAT on screening cells (pooled) 3+ 3+ 3+ 2+ 2+ 1+ 0 0 0 0

Panel of Group O Cells

rr rr r′r R1R1 R1R1 R2R2 R2R2 r″r″

Undiluted serum 3+ 3+ 3+ 3+ 3+ 3+ 3+ 3+
Serum diluted 1:64 1+ 1+ 1+ 0 0 1+ 1+ 1+

Agglutination reactions are graded as 1+ to 4+.

381
382 Immune Hemolytic Anemias

The foregoing comments point out that critical aspects


TABLE 10-5. RESULTS AFTER 1-IN-5 DILUTION of managing patients with autoantibodies who require
OF 119 SERA transfusion are to develop a detailed protocol and ob-
tain appropriate reagents and cells for adsorptions in
Antibody(ies) All Cells
Nonreactive Identified Reactive
advance, rather than waiting until the arrival of a patient
with AIHA who has an urgent need for transfusion.
Warm autoantibodies only 20 (22%) 73 (78%) Technical information regarding adsorption proce-
Warm autoantibodies + 7 (27%) 5 (19%) 14 (54%) dures have been published,48,68 and further details are
alloantibodies provided in Chapter 6.
From Leger RM, Garratty G: Evaluation of methods for detecting alloantibodies
underlying warm autoantibodies. Transfusion 1999;39:11–16.
WARM AUTOADSORPTION TECHNIQUE
The optimal adsorption technique, when feasible, is
in the procedure. The use of PEG for differential
warm autoabsorption. With this technique, one absorbs
adsorption procedures has been proposed by several
the autoantibody from the patient’s serum at 37°C using
groups of investigators.53,59,61,62 There is general agree-
the patient’s own RBCs after first eluting some of the
ment that the use of PEG reduces the time required for
autoantibody. The procedure can be modified by
allogeneic adsorptions and is as effective as using
enzyme treatment of the patient’s RBCs after the elution
ZZAP-treated RBCs.53
procedure.47 Alternatively, one can use ZZAP reagent,
Some precautions and qualifying statements about
which dissociates IgG from RBC and simultaneously
the use of PEG have also been made. Some have sug-
results in their treatment with a proteolytic enzyme.48
gested that weak alloantibodies cannot be detected
After adsorption of the autoantibodies, the serum can
using PEG,60,63-66 whereas in other studies this has not
then be tested for alloantibodies, as alloantibodies will
been the case.53,59,62 Further, some of these differences
not be adsorbed onto the patient’s own RBCs.
of opinion might be because the methods described
It has been our experience that most autoantibodies
have varied among investigators regarding such
have IAT titers of less than 16 with saline suspended
details as the source and composition of PEG and the
RBCs, and two autoadsorptions often removes all
serum:PEG:RBCs ratio that is used.
autoantibody present in the serum. If the autoantibody
Finally, the savings in time when using PEG rather
titer is higher than 16 by the IAT, more autoadsorp-
than ZZAP is, at least in part, lost by the more exten-
tions could be necessary to remove all autoantibody.
sive phenotyping that is necessary for selecting RBCs
An even simpler rule of thumb is as follows: If the
to be used for the adsorptions.46 ZZAP denatures anti-
IAT is 1+, one adsorption is usually all that is required;
gens in the Kell and Duffy systems as well as S, and
if the IAT is 2+, two adsorptions should be performed;
substantially denatures s so that one need not be con-
for a 3+ IAT, three adsorptions are likely to be needed.
cerned about the phenotype of adsorbing cells regard-
When the IAT is 4+, three or more adsorptions are gen-
ing these antigens.48 It might be less labor intensive to
erally necessary to remove all autoantibody.
determine the Rh, Kell, and Kidd (and possibly Duffy
Following the autoadsorptions, the adsorbed serum
and Ss) system phenotypes of the patient (which
is retested. It is recommended that potentiators such
should be done in any case) and to minimize the
as PEG (which enhances autoantibody reactivity) are
number of RBCs used for adsorptions than to
not used at this stage. If a negative reaction is
routinely use multiple (e.g., three) samples of RBCs
obtained, it is assumed that all the serum reactions
for adsorptions. Indeed, if one determines the Rh,
were due to the autoantibody. If a positive reaction
Kell, and Kidd system phenotypes of the patient, it is
still occurs, the adsorbed serum should be tested
often a simple matter to select one or two examples of
against a panel of RBCs to determine whether alloan-
RBCs that would be appropriate for allogeneic
tibody is present or whether autoantibody is still
adsorptions.40,46,59
present, thus requiring further autoadsorptions.
In planning ahead for management of patients who
In some instances, not all serum antibody is removed
will need allogeneic adsorptions, large volumes of
even after multiple adsorptions.54 This could be
phenotyped RBCs may be obtained, treated with
because the autoantibody is so strong that further
ZZAP or ficin if desired, and stored in the liquid state
adsorptions are necessary or because the autoantibody
in Alsever’s solution, ACD, or CPD. If the cells are to
might be reactive against antigens that are destroyed
be treated with ZZAP reagent, one merely needs to
by enzymes or ZZAP. If the latter is the case, then
select R1R1, R2R2, and rr cells, one of which is Jk(a–)
adsorptions with untreated RBCs may be necessary.
and one Jk(b–). Experience in reference laboratories
indicates that RBCs for allogeneic adsorption proce-
dures can be stored in the liquid state for about one to WARM AUTOADSORPTION TECHNIQUE FOR
two months, with or without prior treatment with RECENTLY TRANSFUSED PATIENTS
ZZAP or proteolytic enzymes. Alternatively, if the
need for them is infrequent, they can be glycerolized We have long recommended that the warm autoad-
and cryopreserved in appropriate sized aliquots.40,67 sorption technique not be used in recently transfused
Blood Transfusion in Autoimmune Hemolytic Anemias 383

patients; it seems possible that donor RBCs that are evidence that the autoadsorption procedure should
still circulating could adsorb both alloantibody and not be used in recently transfused patients.
autoantibody from the patient’s serum in vitro.69 Adsorptions using allogeneic RBCs should be used for
There were no published data to substantiate such an patients who have received transfusions within the
opinion, however, and the percentage of a minor pop- last 3 months.70
ulation of RBCs that would invalidate an autoadsorp-
tion procedure was unknown. DETERMINING THE TIME NEEDED FOLLOWING A
To resolve this question, Laine and colleagues70 per- TRANSFUSION BEFORE AN AUTOADSORPTION
formed in vitro experiments in which D, E, K, Fya, and PROCEDURE IS APPROPRIATE
Jka antibodies were absorbed with mixtures of
antigen-positive and antigen-negative RBCs to deter- Some investigators have suggested that it is possible
mine the lowest concentration of antigen-positive to determine whether an autoadsorption would be
RBCs capable of removing all alloantibody reactivity. accurate in a recently transfused patient by perform-
The percentage of antigen-positive RBCs in each ing antigen typing to see whether transfused cells
mixture was determined using flow cytometry. were still circulating. Laine and associates70 studied in
Table 10-6 shows the breakpoints of the percentages vitro mixtures of RBCs to determine whether small
of antigen-positive RBCs at which antibody was and amounts (2%–6%) of “transfused” RBCs could be
was not detected after three adsorptions with the detected directly by antigen typing. Small percentages
mixtures. Small amounts (2%–6%) of antigen-positive of RBC positive for E, K, and Jka could not be detected
RBCs completely removed anti-D, -E, and -Fya. with monoclonal reagents by macroscopic reading
Reactivity of two examples of anti-K was removed by (Table 10-7). Thus, if antigen typing was used to look
11% and 17% of K+ RBCs, respectively. Anti-Jka reac- for the presence of circulating transfused RBCs, the
tivity was completely removed by 4%–5% of Jk(a+) results could be misleading. This procedure should
RBCs using a PEG adsorption, although not by 11% of not be relied on to determine whether autoadsorption
Jk(a+) RBC in a ZZAP absorption procedure. The would be “safe.”
preadsorption titers for all antibodies were not Although Laine and associates70 determined that an
significantly different from the titers after adsorption autoadsorption test is not indicated following a recent
with antigen-negative RBCs. transfusion, their study does not provide insight into
The authors concluded that small amounts of how far removed from transfusion such a test could
antigen-positive RBCs are generally capable of remov- be done. In healthy subjects, RBCs live for about
ing all alloantibody reactivity, thus providing scientific 110–120 days.71 For a patient who has autoantibodies

TABLE 10-6. ANTIBODY TITRATION RESULTS AFTER ADSORPTION WITH DIFFERENT


PERCENTAGES OF ANTIGEN-POSITIVE RBCs

Antigen-Positive Percentage of Antibody Titer


Antibody RBCs Antigen-Positive RBCs after Adsorption

ZZAP PEG
Anti-D R,r 0 8 8–16
1 0–1 0–2
2 0 0
Anti-E E+e– 0 4 16
1 1* 0–1
3 NT 0
Anti-K
Example 1 K+k+ 0 NT 32
13 NT 1
17 NT 0
Example 2 K+k+ 0 NT 32–64
10 NT 1
11 NT 0
Anti-Fya Fy(a+b–) 0 NT 64
5 NT 2
6 NT 0
Anti-Jka Jk(a+b–) 0 32 32–64
4 4 0–1
5 8 0
11 4* NT

* Higher percentage of antigen-positive RBCs not used for adsorption studies.


From Laine EP, Leger RM, Arndt PA, Calhoun L, Garratty G, Petz LD: In vitro studies of the impact of transfusion on the detection of
alloantibodies after autoadsorption. Transfusion 2000;40:1384–1387.
384 Immune Hemolytic Anemias

Toy72 described simple and rapid techniques for pre-


TABLE 10-7. DETECTION OF ANTIGEN-POSITIVE serving RBCs using PVP-methanol or formaldehyde
CELLS BY USING MONOCLONAL AND fixation. Using these methods, RBCs could be stored
POLYCLONAL REAGENTS for six months and, when used for autoadsorption, the
results were comparable to those using fresh cells.
Percentage of Monoclonal Reagent
Antigen-Positive (Direct Polyclonal
Antibody RBCs Agglutination) Reagent (IAT) ALLOGENEIC ADSORPTION
Anti-E 100 4+ NT As indicated previously, the warm autoadsorption
11 1+mf* NT test is not feasible if a patient has been transfused
6 0 NT
3 0 NT recently or if an adequate volume of the patient’s pre-
2 0 NT transfusion RBCs is not available. In either of these
Anti-K 100 4+ 4+ circumstances, the optimal procedure is the allo-
10 0 Micro† geneic adsorption technique—adsorption of autoanti-
5 0 Micro
2 0 0
body from the patient’s serum using allogeneic RBCs
1 0 0 of varying phenotypes.
Anti-Jka 100 3+ 3+ For example, performing an adsorption using a
9 0 1⁄ +mf
2 Jk(a-) cell of a serum containing a warm autoantibody
6 0 Micro and an anti-Jka removes the autoantibody but not the
2 0 0
anti-Jka. As with the warm autoadsorption technique,
* Mixed field. the allogeneic RBCs can be enzyme- or ZZAP-treated.
† Microscopic.
The major practical limitation to the use of the allo-
From Laine EP, Leger RM, Arndt PA, Calhoun L, Garratty G, Petz LD: In vitro
studies of the impact of transfusion on the detection of alloantibodies after geneic adsorption technique is obtaining an adequate
autoadsorption. Transfusion 2000;40:1384–1387. supply of RBCs of appropriate types for the adsorp-
tion (several mL of packed RBCs of each type are
needed).
without hemolytic anemia, in whom transfused and We suggest using three samples of allogeneic
autologous RBCs would be expected to have normal RBCs—one rr, one R1R1, and one R2R2; one sample
survival, waiting for 3 to 4 months after a transfu- should be Jk(a-) and one should be Jk(b-). We prefer to
sion before doing autologous adsorption would be use ZZAP-treated RBCs, as these will not adsorb any
appropriate. antibodies to any antigens in the MNS, Kell, Lutheran,
In patients with AIHA, transfused RBCs can be and Duffy systems but will adsorb autoantibodies
expected to have a shortened survival time.71 efficiently. On rare occasions, however, autoantibodies
Therefore, autoadsorption would seem appropriate at have specificities that are directed against antigens
a time interval much shorter than 3 months for destroyed by ZZAP (e.g., Ge, Ena, Kell system anti-
patients with severe hemolysis and a markedly abnor- gens). Thus, if one is unsuccessful after many adsorp-
mal RBC survival time who need repeated transfu- tions, one should consider adsorbing with untreated
sions. The significance of this finding, however, is RBCs. The adsorption procedure using allogeneic
negated by the fact that such patients are also likely to RBCs is similar to that for using autologous RBCs. By
need repeated transfusions at short intervals. An accu- observing the pattern of reactivity left in the serum in
rate determination of the time when all allogeneic relationship to the phenotype of the adsorbing RBCs,
cells have been eliminated is not feasible. one can determine the specificity of the alloantibody.
Although waiting for 3 months after transfusion Disadvantages of adsorbing with allogeneic RBCs
before allowing an autoadsorption would seem to be are that large volumes of phenotyped RBCs are needed,
a conservative approach, it might be the most appro- and that alloantibodies to high-frequency antigens will
priate policy. Accordingly, the allogeneic adsorption be adsorbed by all three RBC samples, leading to a false
technique is generally the optimal method for detect- assumption that no alloantibody is present. Never-
ing alloantibodies for patients with WAIHA who need theless, allogeneic adsorption provides the greatest
repeated transfusions. safety of any procedure other than autoadsorption for
selection of blood. Although the possible presence of
STORAGE OF AUTOLOGOUS CELLS FOR alloantibodies of some specificities is ignored, the prob-
FUTURE AUTOADSORPTIONS ability of an alloantibody-induced hemolytic transfu-
sion reaction is minimal.
Because the warm autoadsorption technique is so Transfusion services should plan ahead to circum-
useful, we recommend storing some of the patient’s vent the problem of availability of cells for adsorption,
RBCs that are obtained before the first transfusion as indicated previously.
episode so that they can be used in future autoab- In some situations, fewer samples of adsorbing RBCs
sorptions in the event that continued transfusions are may be required. Because the patient’s extended phe-
required.69 The RBCs may be stored in ACD, CPD, or notype should be determined prior to the first trans-
the frozen state if facilities are available. Reid and fusion, one may select RBCs for adsorption on the
Blood Transfusion in Autoimmune Hemolytic Anemias 385

basis of these results. For example, if the patient is RBCs lacking the more strongly reactive antigen
R1R1, K-negative, Jk(a-), Jk(b+), one may simply survive significantly better than RBCs that do contain
adsorb with ZZAP-treated RBCs that are R1R1 and it. The dilution technique may be used for determin-
Jk(a-). It could be less labor intensive to determine the ing Rh-relative specificity. In essence, one need only
Rh, Kell, and Kidd system phenotypes of the patient titrate the patient’s serum or eluate against R1R1, R2R2,
and to minimize the number of RBCs used for adsorp- and rr RBCs. Table 10-8 shows results of an eluate that
tions than to routinely use multiple samples of RBC would be interpreted as showing “relative specificity”
for adsorptions. But because phenotyping for all rele- against the e antigen. Such reactions should be
vant antigens might not be feasible for a patient who confirmed by testing against several examples of
has a positive DAT, it is prudent to have available a RBCs with and without the appropriate antigen
supply of the three examples of RBCs previously sug- before making clinical decisions based on the “relative
gested, which can be used for adsorption regardless of specificity” of the autoantibody.
the patient’s RBC phenotype.
SIGNIFICANCE OF AUTOANTIBODY SPECIFICITY
Autoantibody Specificity
Several investigators have studied the in vivo survival
Defining the specificity of the autoantibody73,74 is not of RBCs of varying Rh phenotypes among patients
as important as excluding the presence of alloantibo- who have warm autoantibodies with Rh “specificity.”
dies, but if time allows and if blood lacking the puta- In most instances, detailed serologic data are not
tive antigens can be found expeditiously, it might given, and the autoantibodies are likely to have
promote the survival of transfused RBCs. If the demonstrated “relative specificity.”
autoantibody shows a well-defined specificity (e.g., Mollison75,76 described a case in which survival of
anti-e), compatible blood should be obtained unless the patient’s own e-positive RBCs was shortened
this would delay transfusion significantly. markedly, whereas transfused e-negative RBCs sur-
vived almost normally. The patient’s serum contained
AUTOANTIBODIES WITH RH SPECIFICITY an autoantibody that reacted preferentially with e+
OR “RELATIVE SPECIFICITY” RBCs (Fig. 10-1).
Salmon77 described two patients who had anti-e and
Serologists are frequently vague regarding the criteria anti-nl autoantibodies. In the first case, the T50 of 51Cr-
used to report an autoantibody as having Rh labeled RBCs was 23 days for -D-/-D- RBCs (e-,
specificity. Most warm autoantibodies react with all nl-), 24 days for cDE/cDE RBCs (e-, nl+), and 12.5 days
RBCs of common Rh phenotypes, but they might fail for cde/cde RBCs (e+, nl+). In the second case, the T50
to react with gene deletion cells, such as Rhnull cells. was 14 days for cDE/cDE RBCs but only 4 days for
Other autoantibodies react with all RBCs tested but CDe/cde RBCs.
react to a higher titer or score against RBCs bearing a von dem Borne and colleagues78 reported on a
particular Rh antigen. In either case, the autoantibody patient who had autoanti-e and anti-nl antibodies.
is usually said to have Rh specificity without distin- The 51Cr half-time of CDe/CDe RBCs was 1.9 days,
guishing such reactions from each other or from the and that of cDE/cDE RBCs was 4.0 days.
clear-cut specificity of Rh alloantibodies, wherein cells In Höllander’s patient, the autoantibodies had anti-
lacking the appropriate antigen yield strictly negative D specificity; whereas cde/cde blood survived for at
reactions. We will use the term relative specificity to least 31 days, CDe/cde blood survived for only 3
refer to antibodies that react with all normal RBCs days.79 In Crowley and Bouroncle’s patient, two auto-
bearing common Rh antigens but react consistently to antibodies—anti-D and anti-E—were present, and
a higher titer or score against RBCs containing one or cde/cde cells survived normally.80 In the patient of
another Rh antigen. Wiener, Gordon and Russow,81 the autoantibody
Tests for determining “relative specificity” of reacted to highest titers with cells containing the rh’
autoantibody should be performed if time allows, as (C) factor; when transfused with blood lacking this

TABLE 10-8. ELUATE SHOWING ANTI-E “RELATIVE SPECIFICITY”

Dilutions of Eluate

2 4 8 16 32 64 128 256

rr(cde/cde) 4+ 3+ 3+ 2+ 2+ 1+ 0 0
R1R1(CDe/CDe) 4+ 3+ 3+ 2+ 2+ 1+ 0 0
R2R2(cDE/cDE) 3+ 2+ 1+ 0 0 0 0 0

Agglutination reactions are graded as 1+ to 4+.


386 Immune Hemolytic Anemias

100 generally made if it would be necessary to give


D-positive blood to a D-negative patient.
In contrast to our approach, some immunohemato-
logists recommend ignoring the specificity of the
75 autoantibody. This recommendation is based on two
considerations. First, the evidence indicating good sur-
Percentage of survival

vival related to autoantibody “relative specificity” is


not extensive. Indeed, in some reports no difference in
50 survival in vivo can be demonstrated,85 and in others,
the benefit has been minimal.78 In other instances,
good survival of donor blood lacking the more reactive
antigen has not been shown to be due to the autoanti-
body specificity, as survival of transfused RBCs con-
25
taining the more reactive antigen has not been
studied.29,80,83,84 Second, if one transfuses RBCs that
lack an antigen with which an autoantibody reacts,
one might need to use RBCs containing an antigen not
0 found on the patient’s own RBCs, thus causing the
0 10 20 30 potential for alloimmunization. This does not seem to
Days be a critical argument, as typing for Rh antigens other
FIGURE 10-1. Survival, in a ddccee patient with autoimmune than the D antigen is not part of routine blood transfu-
haemolytic anaemia of e+ (DCCee) red cells (•), estimated by differen- sion practice. Such a precaution might be warranted,
tial aggultination, and of e– (DccEE) cells (×), estimated by 51Cr-labeling however, as some data suggest that patients with
and corrected for Cr elution. The patient’s serum contained an
autoantibody reacting preferentially with e+ cells. (From Mollison PL:
AIHA have an increased incidence of development of
Measurement of survival and destruction of red cells in haemolytic RBC alloantibodies after transfusion (see the earlier
syndromes. Brit Med Bull 1959;15:59.) section on the incidence of alloantibodies in patients
with AIHA who require transfusion).

factor, the patient made a complete and lasting recov-


ery. Previously, she had been treated with randomly “LEAST INCOMPATIBLE UNITS”
selected Rh-positive donors and had failed to
improve. Ley, Mayer, and Harris’s82 patient was group The term least incompatible unit is not an official term
0 cde/cde. RBCs of phenotype cDE/cDE survived in transfusion medicine and is not defined in the
normally (51Cr T50 = 25 days), but cde/cde cells sur- medical literature.86 It appears to mean the selection
vived even less well than the patient’s own cells (51Cr of a unit of blood that gives weaker reactions in the
T50 5 days and 13–14 days, respectively). compatibility test than other incompatible units. That
Hogman, Killander and Sjolin’s case was a 13-year- is, one may perform cross-match tests using a number
old child of phenotype CDe/CDe who had formed of donor units that are ABO- and Rh-matched with the
autoanti-e antibody and an apparent “nonspecific” patient and then select the one that reacts least
component.83 The latter component did not appear to strongly. The rationale for using “least incompatible”
be of much importance, as cDE/cDE RBCs survived units appears to be that the stronger reactions could
normally. be caused by an alloantibody. The use of this term
Bell and coworkers84 mentioned one patient with apparently lingers on from the days before effective
anti-e who tolerated two e-negative units with the and practical serologic tests were devised for the
expected rise and maintenance of hemoglobin levels. identification of alloantibodies in the presence of
No further details are given. autoantibodies that react with all RBCs.
Habibi and associates29 transfused cDE/cDE RBCs Selecting “least incompatible” units must not be
to six patients with autoantibodies of e, ce, or Ce considered an acceptable alternative to the techniques
specificities. The blood “proved normally efficient in described earlier in this chapter for selecting donor
vivo,” but two of six homozygous e+ patients devel- units for transfusion. Reliance on “least incompatible”
oped anti-E. units instead of performance of appropriate serologic
Although the preceding data are scanty, we feel that studies is a dangerous practice and should be aban-
if an autoantibody demonstrates “relative specificity” doned, except in extremely urgent settings when time
(e.g., if the titer against RBCs containing the e antigen to perform adequate serologic tests is insufficient.
is consistently two tubes higher than when tested Some transfusion services appear to use the term
against cells lacking the e antigen), it is preferable to “least incompatible unit” in another context. They
avoid transfusion of blood containing the antigen in select donor units for transfusion after adequately
question, even though this could involve the deliber- accounting for RBC alloantibodies or select units on
ate administration of RBCs containing Rh antigens the basis of extended phenotyping. Nevertheless, any
that the patient lacks. An exception to this course is unit selected is incompatible with the patient’s
Blood Transfusion in Autoimmune Hemolytic Anemias 387

autoantibody. Transfusion services might choose to though the donor blood appears to be compatible, the
cross-match the selected units with the patient’s transfused RBCs cannot be expected to survive nor-
serum, although all such cross-matches are incompat- mally. Indeed, Mollison71 has stated that in all those
ible. Choosing the “least incompatible unit” from conditions in which a hemolytic anemia is due to
among those that have been selected might provide a some extrinsic mechanism rather than to any intrinsic
level of comfort to the transfusion service personnel RBC defect, transfused normal RBCs are expected to
and can do no harm, but this provides no known undergo accelerated destruction.
benefit to the patient. Some variability in reactivity
caused by an autoantibody can be expected to occur Selection of Blood: Summary
when a number of units are cross-matched with the
patient’s serum; this phenomenon is due simply to the With regard to selection of blood for transfusion to
limitations of precision of serologic reactions. patients with WAIHA, we feel that it is most impor-
The term “least incompatible” unit should be tant to determine the extended RBC phenotype of the
placed in the garbage heap of serologic terminology. It patient, to save some of the patient’s RBCs for future
is not defined in transfusion medicine nomenclature; warm autoadsorption tests, to compare the strength
it is undoubtedly used differently by various transfu- of the DAT and IAT, to test for antibody specificity
sion services; its use does not convey information using a routine RBC panel, and to perform the warm
regarding the extent of compatibility testing per- autoadsorption test. In a recently transfused patient,
formed; and finally, the term implies that this is an an allogeneic adsorption test should replace the
acceptable alternative to adequate serologic evalua- warm autoadsorption test unless pretransfusion
tion prior to transfusion of patients with AIHA.86 RBCs are available. If time allows, one may also test
for autoantibody specificity (e.g., Rh “relative specifi-
city”). Table 10-9 summarizes our recommendations.
AUTOIMMUNE HEMOLYTIC ANEMIA WITHOUT
SERUM AUTOANTIBODY
The Optimal Frequency of Tests for
In contrast to the previously described problems fre- Alloantibodies in a Patient with AIHA
quently encountered among patients with AIHA, it Who Is Transfused Repeatedly
should also be pointed out that in some patients, the
autoantibody does not interfere with compatibility The question frequently arises as to how often a search
testing. This is true because the autoantibody might for alloantibodies using adsorption procedures must
be undetectable in the patient’s serum (apparently be performed in a patient with AIHA who is being
because it is entirely adsorbed onto the patient’s transfused repeatedly. As pointed out by Leger and
RBCs) or is detectable only by techniques more sensi- Garratty,53 patients who have autoantibodies should
tive than are used in routine compatibility tests. Even receive the same protection from hemolytic transfusion

TABLE 10-9. SUMMARY OF METHODS USED IN SELECTION OF BLOOD FOR TRANSFUSION TO PATIENTS WITH
WARM ANTIBODY AUTOIMMUNE HEMOLYTIC ANEMIA*

A. Patient not recently transfused (within previous 3 months)


1. Determine patient’s ABO and Rh phenotype.
2. Determine the patient’s extended RBC phenotype (e.g., K, Jka, Jkb, Fya, Fyb, S), if feasible.
3. Compare strength of DAT and IAT. If IAT is stronger, the presence of an alloantibody is highly suspected.
4. Test patient’s serum against a panel. If an alloantibody causes stronger reactions than the autoantibody, the alloantibody specificity might
be evident.
5. Obtain RBCs for autoadsorption tests; save as many RBCs as practical in anticoagulant or in frozen state for use in subsequent warm
autoadsorption tests, should repeated transfusions be required.
6. Perform autoadsorptions for detection of alloantibodies.
7. If RBCs are not available for the autoadsorption test, perform allogeneic adsorptions for detection of alloantibodies.
8. If the IAT is strongly reactive and adsorptions are not possible (lack of time or lack of autologous or allogeneic RBCs), prepare a dilution
of the patient’s serum that reacts about 1+ by IAT and test against a panel.
B. Patient recently transfused (within previous 3 months)
1. Pretransfusion RBCs are available for warm autoadsorption test.
a) Follow steps 1–5, above (accurate determination of the patient’s RBC phenotype could be difficult or impossible)
b) Perform warm autoadsorption for detection of alloantibodies, using pretransfusion RBCs.
2. Pretransfusion RBCs are not available for warm autoadsorption test.
a) Follow steps 1–4, above (accurate determination of patient’s RBC phenotype could be difficult or impossible)
b) Perform allogeneic adsorptions for detection of alloantibodies or, if extended RBC phenotype of the patient is known, one may use
phenotypically matched RBC units, if available. Using partially matched units (e.g., units matched for Rh and K) will provide only
partial safety (see text).

* If a patient has never been transfused or pregnant, it is highly unlikely that clinically significant alloantibodies are present.
Testing the patient’s autoantibody for specificity could provide additional benefit (see text).
388 Immune Hemolytic Anemias

reactions as other patients. In particular, the American Jkb, Fya, Fyb, S, and s antigens), and all donor units
Association of Blood Banks’ Standards indicate that if must be matched with the patient’s. If the patient’s
a patient has received a transfusion or been pregnant RBCs can be phenotyped for all of these antigens, and
within the preceding 3 months, the sample must be if the blood supplier can provide units that are nega-
obtained from the patient within 3 days of the sche- tive for all of these antigens not present on the
duled transfusion.87 Indeed, Shulman and colleagues88 patient’s RBC, this method of selecting blood would
have published data indicating that 13 of 60 retrospec- appear to be about as safe as using an allogeneic
tively studied patients developed newly detectable adsorption procedure. Although performing an
antibodies within 83 hours of a sample reported to be extended phenotype is a labor-intensive and expen-
negative for the new antibody. Thus, after a patient sive procedure, it need be done only once per patient,
with autoantibodies receives a transfusion, compati- and the information can be used for all subsequent
bility test procedures, including adsorption studies, transfusions. This might not be a significant advan-
should be performed on samples obtained within tage, however, as data from the Los Angeles Red
3 days of subsequent transfusions. Cross (LARC) Reference Laboratory (unpublished)
shows that only 90 of 418 patients (22%) needed more
The Use of Phenotypically Matched than the initial set of adsorptions, evidently because
RBCs for Transfusion the patients responded to therapy and repeated trans-
fusions were not required. Further, one must keep in
As the frequent performance of adsorptions is labor mind that even in the most skilled hands, extended
intensive, there is a never-ending search for less techni- phenotyping of RBCs that have a strongly positive
cally demanding but safe approaches to providing direct antiglobulin test might not be possible. Indeed,
blood for patients with AIHA. Some investigators Shirey and coworkers89 could not obtain a reliable
suggest the transfusion of RBCs that are prophylacti- phenotype from 40% of their patients, and in these
cally antigen matched (PAM) with the patient’s, rather cases, adsorption procedures were necessary for
than performing adsorption studies to detect and iden- alloantibody detection and identification.
tify alloantibodies.89 Providing phenotypically matched Data from the LARC Reference Laboratory91 sug-
RBCs can provide a significant measure of safety,90 but gests that the use of phenotypically matched RBCs
some caveats and precautions should be noted. prevents most of the alloantibody-induced hemolytic
Performing an autoadsoption provides protection transfusion reactions that potentially would occur in
against the presence of antibodies against high- patients with AIHA. Table 10-10 shows the specificities
incidence antigens, whereas providing blood selected of 202 alloantibodies detected in the sera of 418
on the basis of the patient’s phenotype or by the patients with AIHA. Anti-E was by far the most
alloadsorption technique does not. Therefore, autoad- common alloantibody detected, being twice as
sorption should be considered a preferable approach common as the next most common specificity, anti-K.
when feasible. The next most common group included anti-C, -Fya,
To provide adequate safety, the patient’s extended -Jka, and anti-c, in that order. A third group of alloanti-
phenotype must be determined (D, C, E, c, e, K, Jka, bodies—anti-Jkb, -S, -D, -e, “HTLA”, -M, -V, -Jsa, -Cw,

TABLE 10-10. ALLOANTIBODY SPECIFICITIES OF 202 ANTIBODY-CONTAINING SERA FROM 418 AIHA PATIENTS

Specificity* n Percentage Present in Antibody-Containing Sera Percentage Present in Total AIHA Patients

Anti-E 92 46 22
-K 45 22 10.8
-C 36 18 8.7
-Fya 30 15 7.8
-Jka 21 10 5.0
-c 20 10 4.8
-Jkb 19 9.4 4.6
-S 17 8.4 4.0
-D 14 7 3.4
-e 10 5 2.4
“HTLA” 9 4.5 2.2
-M 9 4.5 2.2
-V 8 4 1.9
-Jsa 7 3 1.7
-Cw 7 3 1.7
-Lea 7 3 1.7
-Wra 5 2.5 1.2
-s 4 2 1.0
-rhi 4 2 1.0

* Other specificities detetected in only 1 or 2 sera included anti-Kpa, -VS, -P1, -Ch, -G, -N, -Dia, -Ce, -KnMca, -U, -Lua, -Lu14, -He, -Xga, -Mit, -Fy5.
From Garratty G, Petz LD: Approaches to selecting blood for transfusion to patients with auto-immune hemolytic anemia. Transfusion 2002;42:1390–1392.
Blood Transfusion in Autoimmune Hemolytic Anemias 389

Lea, -Wra, -s, and –rhi—were present in 1% to 10% of compatibility test strictly at 37°C and to use only
the alloantibody containing sera. A fourth group of saline-suspended RBCs (i.e., without potentiators).
various specificities was associated with only one or Cold agglutinins from only 7% of patients with cold
two of the patients. Approximately 40% of the sera agglutinin syndrome react at 37°C using saline-sus-
contained alloantibody of only a single specificity; 30% pended RBCs, although we found positive reactions
contained two specificities; and 16% contained three in 30% of cases in albumin media.94,95 If positive reac-
specificities. Approximately 10% of the alloantibody- tions occur at 37°C with saline-suspended RBCs, one
containing sera contained alloantibodies of four or must first suspect faulty technique. If cells and serum
more specificities. are not prewarmed before mixing, if centrifugation is
Providing PAM RBCs might present a significant performed at a temperature lower than 37°C, or if the
problem for hospitals and smaller blood suppliers. initial washes of the cells after incubation do not use
The phenotypes of the 12 patients who received PAM saline at 37°C, reactions can occur within seconds.
RBCs (listed in Table 2 of Shirey and associates89) Even if direct agglutination is not evident, comple-
range from 0.0002 to 0.09 (mean = 0.04), meaning that ment might be bound by the antibody reactivity and
51,000–65,000, or a total of 76,300 random units result in a positive IAT using polyspecific antiglobu-
would have to be screened to obtain the 149 PAM lin serum (one molecule of IgM antibody might bind
RBCs that were transfused to these patients. Even if several hundred molecules of complement). This
the frequency of the rarest of the phenotypes, E–, K–, reaction may be circumvented by using anti-IgG
Fy(a–b–), S– (patient #9, who was probably African antiglobulin serum.
American) is removed from the calculation (as this The advantages of this method are that time-
phenotype could be obtained more easily by screen- consuming autoadsorptions of the patient’s serum are
ing African Americans), 51–6714, or a total of 11,306 not necessary and that the method can be used even if
random donors would have to be screened to obtain the patient has recently received a transfusion.
the 136 PAM RBCs transfused to 11 patients.91 Several disadvantages are also apparent. First, it is
Nevertheless, Lau and colleagues90 have suggested obvious that RBC alloantibodies reacting at tempera-
that, with the use of a sophisticated computer tures lower than 37°C will not be detected. This
program, it is feasible and cost effective in a rela- finding is of little consequence, as alloantibodies that
tively homogeneous population in Hong Kong to do not react in vitro at temperatures less than 37°C are
obtain phenotype-matched blood for all patients rarely, if ever, clinically significant. Indeed, the
without need for pretransfusion antibody screening. Standards for Blood Banks and Transfusion Services
Partial phenotyping (e.g., for Rh, K and Jka anti- of the American Association of Blood Banks does not
gens) would not provide protection against alloim- require a room temperature incubation phase of the
munization by antigens of other blood group systems crossmatch but instead states that methods for testing
that can cause hemolytic transfusion reactions, and for unexpected antibodies “shall include 37°C incuba-
therefore, it would not preclude the necessity of pre- tion preceding an antiglobulin test using reagent
transfusion adsorption studies.71,92,93 Determining the RBCs that are not pooled.”87
partial genotype of patients’ RBCs using DNA tech- It is also true that potentiator-dependent antibodies
nology might be helpful in identifying at least some will be missed, but here, again, the risk is minimal
RBC antigens (see the earlier discussion of red cell because such antibodies are quite unusual.96 Because
phenotyping and genotyping). compatibility testing at 37°C is quicker than other
Whether implementation of this approach is feas- methods, it can be used even if the patient has
ible and cost effective at many blood centers has not recently received a transfusion, and because it results
been determined. If the intention is to place emphasis in a low risk of missing clinically significant allo-
on providing phenotype-matched units, one must antibodies, we believe it is the method of choice.
determine that the blood supplier could provide such Attention to certain technical details is crucial,
units reliably, and one must recognize that adsorption however, to be certain that one is truly working strictly
studies will be required in cases in which a patient’s at 37°C.
RBCs cannot be phenotyped and/or when the blood One must validate that procedures are actually
supplier cannot provide matched units. being carried out strictly at 37°C. A heated centrifuge
or a centrifuge in a 37°C warm room may be used, a
centrifuge may be placed in an incubator, or the tubes
COMPATIBILITY TESTING IN may be placed in centrifuge cups containing warm
COLD-ANTIBODY AIHAs water. Samples transferred from a 37°C water bath
and centrifuged immediately at room temperature
drop by approximately 7–8°C after only one minute of
Cold Agglutinin Syndrome centrifugation. One must be aware that, if one uses
PERFORMING COMPATIBILITY TESTING AT 37°C saline at 40–45°C, the temperature will drop by a few
USING SALINE-SUSPENDED RBCs degrees when it enters a test tube and by a few more
degrees when centrifuging is in progress. A few
There are several approaches to compatibility testing simple experiments are all that is required to deter-
for patients with CAS. One method is to perform the mine the appropriate conditions.
390 Immune Hemolytic Anemias

COLD AUTOADSORPTION AND Pirofsky and Rosner100 described the use of DTT at
ALLOGENEIC ADSORPTION a concentration of 0.01 M in a rapid 15-minute, 37°C
incubation test system. Dialysis was not required.
An alternative approach is to adsorb the cold autoan- They reported that this procedure caused at least a
tibody from the patient’s serum before performing fourfold or greater decrease in IgM antibody titers
the compatibility test. If facilities are not available to without affecting the activity of IgG antibodies.
work strictly at 37°C, one or two cold autoadsorp- Olson and associates101 used DTT in a concentration
tions will frequently remove enough of the cold of 0.01 M, added equal volumes to test sera, and incu-
agglutinin so that compatibility tests can be per- bated for 30 minutes at 37°C. Thirty sera that con-
formed with less stringent control of the incubation tained RBC antibodies reactive by the IAT showed
temperature. It is interesting that cold autoadsorp- virtually no alteration in activity after DTT treatment,
tions remove antibody reactive at 30–37°C before all while 20 sera containing cold-reactive RBC antibodies
antibody reactive at 4°C is adsorbed, thus making showed almost total elimination of activity. However,
compatibility testing feasible without adhering none of the cold-reactive antibodies tested were
strictly to 37°C temperatures. pathologic high-titer cold agglutinins from patients
Even if the transfusion service can work strictly at with CAS.
37°C, cold autoadsorptions might be necessary in the Freedman and colleagues102 reviewed the optimal
small percentage of patients whose antibody is reac- conditions for the use of sulphydryl compounds in
tive at 37°C, even in saline. If a patient has a very dissociating RBC antibodies. They noted that incuba-
high-titer cold agglutinin, one should not attempt to tion at 37°C with 0.2 M 2ME provided the best con-
remove all of the antibody by adsorption. Doing so ditions for inactivating IgM antibodies. Incubation
would require multiple adsorptions even when still failed to inactivate completely the extremely
enzyme-treated RBCs are being used and is not neces- potent autoanti-I (titer of 1,024,000) that was used,
sary. Table 10-11 shows the results of autoadsorbing a however. False-positive reactions in the IAT using
serum with a cold-agglutinin titer of 2048 (saline) and anti-IgG or anticomplement antiglobulin serum were
8096 (albumin). After three adsorptions for one-half obtained consistently when sera that had been
hour each at 4°C using the patient’s papainized RBCs, treated with 2ME were not subsequently dialyzed.
the serum still reacted strongly (4+ with undiluted Although in many cases dialysis for as short as
serum) at room temperature (25°C), but it no longer 30 minutes was sufficient, in others overnight dialy-
reacted at 37°C. sis was found to be necessary. Incubation of serum
In recently transfused patients, allogeneic adsorp- with DTT produced a slower effect than did incuba-
tion studies can be performed as for warm-antibody tion with 2-mercaptoethanol, and incubation for
AIHA. This is rarely necessary if compatibility tests 2.5 hours was necessary to reduce the anti-I titer
are carried out as described previously. from 1,024,000 to 1024.
Using either reagent, IgM alloantibodies will, of
OTHER METHODS course, be inactivated in addition to the cold autoag-
glutinins. Other disadvantages are that blood bank
An alternative approach to compatibility testing is to technologists are often unfamiliar with the use of such
adsorb the serum with rabbit erythrocyte stroma, reagents, and both reagents can inactivate or diminish
which can be used to adsorb anti-I and –IH40 but complement activity.
might also remove clinically significant alloantibodies
(notably anti-B, -D, -E, and others).97 TRANSFUSION OF ADULT-i RBCs
Still another approach to compatibility testing in
cold agglutinin syndrome is to inactivate the IgM cold Some investigators have suggested that adult-i RBCs
agglutinin with 2-mercaptoethanol (2ME) or DTT.98,99 be used for transfusion of patients with cold agglutinin

TABLE 10-11. ABSORPTION OF COLD AGGLUTININ BY PATIENT’S OWN RBCs AT 4°C

Titer against Adult OI RBCs

4°C 25°C 37°C

Saline Albumin Saline Albumin Saline Albumin

Unadsorbed 2048 8098 1024 8098 0 32


Absorbed × 1 1024 2048 256 1024 0 16
Absorbed × 2 256 256 128 256 0 8
Absorbed × 3 128 128 16 64 0 0
Blood Transfusion in Autoimmune Hemolytic Anemias 391

syndrome who have anti-I autoantibodies. van Landsteiner antibody. The hematocrit temporarily
Loghem and associates103 studied the survival of I and improved from 13.5% to 20% but decreased over the
i RBCs labeled with 51Cr in one patient with chronic next 36 hours to 11.5%. A second incompatible transfu-
CAS. They demonstrated normal survival for the i sion of 130 mL again resulted in a temporary rise in
donor RBCs with greatly shortened survival of both the hematocrit to 24.5% but with a drop to 15% over the
patient’s I RBCs and donor I RBCs. Woll and cowork- next 36 hours. The urine then abruptly became clear
ers104 reported one patient with transient CAS who and at this point, 100 mL of frozen-thawed RBCs from
responded to transfusion of warmed, freeze-thawed a compatible type O, Tj(a-) donor were transfused
adult-i RBCs. These researchers did not test the sur- through a warming coil adjusted to 37°C. Following
vival of adult I RBCs, however. Bell and colleagues84 this transfusion, the hematocrit rose to 20%, and the
reported unfavorable experiences transfusing two patient’s hemolytic anemia stabilized and then gradu-
patients with CAS with adult-i RBCs. Two adult-i units ally resolved. The authors state that these were the first
given to a patient with strong anti-I survived for recorded cases of successful compatible transfusion
approximately the same period of time (i.e., 3 to 4 days) therapy in PCH. One should note, however, that both
as several adult-I units given subsequently. In a second patients’ hemolysis spontaneously subsided soon after
patient with anti-I, no elevation of hematocrit was the transfusions of compatible blood and that one
noted after transfusion of two units of adult-i blood. cannot determine the comparative efficacy of the com-
These authors suggested that the minimal I antigen patible and incompatible transfusions from the data
present on adult-i RBCs seemed sufficient to render provided.
them biologically incompatible. Sabio and associates105 reported on a 41⁄2-year-old
Our own experience indicates that transfusion of girl with PCH who was transfused twice with
adult-I RBCs to patients with chronic CAS usually ABO/Rh (D)-compatible, P+-incompatible RBCs.
results in an appropriate rise in hemoglobin. Unusual These transfusions only worsened the patient’s hemo-
patients might fail to respond to transfusion of adult-I globinuria without increasing the hemoglobin con-
RBCs, but a majority of available data indicate that the centration significantly. Transfusion with RBC of the
use of adult-i cells is not a solution to the problem. In rare p phenotype was attempted successfully with a
patients with chronic CAS, the repeated use of i RBCs rise in hemoglobin and amelioration of symptoms.
is certainly not feasible because of their extreme rarity. The patient was also treated with 2 mg/kg prednisone
“without any effect” following the first transfusions.
Nordhagen and colleagues106 reported on four chil-
Paroxysmal Cold Hemoglobinuria dren with acute PCH who required transfusion in spite
Transfusion of compatible blood for patients with of aggressive therapy with corticosteroids. One patient
paroxysmal cold hemoglobinuria (PCH) might be received three transfusions of P-positive blood during
possible if the rare p or Pk cells are available through the first 3 days of hospitalization. One week later, the
a rare donor file. Although the routine cross-match patient became febrile again, with increasing hemolysis
test could appear to be compatible with other RBCs and a hemoglobin that dropped from 7.3 to 4.6 g/dL.
because the antibody reacts only in the cold (usually He was given another transfusion on the 10th day and
<15°C), there are some suggestions that p or Pk RBCs the last on the 14th day. The last transfusion consisted
will survive better.22,105 of washed, P-negative (pp) erythrocytes, after which
Rausen and colleagues22 reported on two children the patient’s condition stabilized.
with PCH who were severely anemic. The first Patients are likely to require transfusion before
patient, a 4-year-old boy with blood type A1, was RBCs of the p phenotype are available. Generally,
transfused with two 100-mL aliquots of frozen- transfusion of RBCs of common P types should be
thawed RBCs from a compatible type O, Tj(a-) donor provided, as patients with PCH often have severe
5 days after the sudden onset of hemolytic anemia. hemolysis, and waiting for availability of p or Pk RBC
The blood was administered at room temperature, is likely to delay a needed transfusion. Successful
and the hematocrit level rose from 11% to 19% follow- transfusion of patients with PCH has been reported
ing the transfusions. Although the hematocrit had by numerous authors and, almost certainly, the trans-
been expected to reach 28%, the authors determined fusions were of P-positive blood.33,106-112
that the post-transfusion blood contained fewer than Although there are some reports of Donath-
5% type A RBCs and therefore concluded that the lack Landsteiner antibodies having other specificities,
of an appropriate rise in the hematocrit was because these are extraordinarily rare (see Chapter 5).
the patient’s own cells were rapidly being hemolyzed.
Within 2 more days, hemoglobinuria subsided, and OPTIMAL VOLUME OF BLOOD
the patient’s hemoglobin rose progressively.
The second child, a 21⁄2-year-old boy with blood type TO BE TRANSFUSED
AB, was treated with intravenously administered
hydrocortisone on the first day of hospitalization, and The optimal volume of blood to be transfused to
subsequently he was transfused with 100 mL of type patients with AIHA varies with the clinical setting. In
AB RBCs that were incompatible with the Donath- patients who have severe hemolysis but who might
392 Immune Hemolytic Anemias

require transfusion only temporarily until therapy Chaplin113 indicates that the most common cause of
becomes effective, the transfusion of modest volumes post-transfusion hemoglobinuria in AIHA might not
of RBCs just sufficient to maintain a tolerable hema- be alloantibody-induced hemolysis but rather the
tocrit level appears advisable. In patients with severe quantitative effect of transfusion in increasing the
anemia, however, there is a tendency to transfuse RBC mass subjected to ongoing autoantibody-
large volumes of blood to restore the hemoglobin and mediated destruction. Such marked post-transfusion
hematocrit to near-normal levels quickly. This hemoglobinemia and hemoglobinuria have the poten-
approach can lead to tragic consequences. tial for a significant degree of associated morbidity
Indeed, Rosenfield and Jagathambal37 point out that and possibly, mortality. Indeed, Gürgey and cowork-
the salutary effect of just 100 mL of packed RBCs can ers114 pointed out that two children who were treated
be quite remarkable when given to a patient with car- with exchange transfusions died of profound hemoly-
diopulmonary embarrassment from anemia. They sis shortly after the procedure (although Heidemann
suggest that 100 mL may be given as needed (perhaps and colleagues115 reported successful exchange trans-
twice daily, depending on the severity of hemolysis) fusion in one patient).
and that there is generally no need to increase the Patients undergoing severe post-transfusion intra-
hemoglobin even to a level of 8 g/dL. The aim of such vascular hemolysis can develop disseminated intravas-
transfusions is to supply just enough RBCs to prevent cular coagulation (DIC), possibly as a result of
hypoxemia while avoiding dangerous reactions procoagulant substances present in RBC lysates.36,116
resulting from overtransfusion. These potential problems emphasize the need for
restraint in the volume of blood transfused per trans-
Complications of Aggressive Transfusion fusion episode to patients with AIHA.
Therapy in Patients with AIHA
CASE SUMMARIES
VOLUME OVERLOAD
An example of such a complication of transfusion
The dangers of overtransfusion among patients with therapy was reported by Bilgrami and associates.116
AIHA are several. If the anemia is very severe (hema- They described a patient who was infected with
tocrit 10–15%, hemoglobin 3.5–5.0 g/dL), and espe- human immunodeficiency virus (HIV) who had
cially if the patient is elderly or if cardiac reserve severe AIHA. His hemoglobin was 2.9 g/dL; hema-
might be reduced, transfusion can overload the circu- tocrit, 8%; corrected reticulocyte count, 1.2%; white
lation easily and precipitate cardiac failure. In slowly cell count, 12,500/μL; platelet count, 101,000/μL;
developing anemia, total blood will not be as severely total bilirubin, 3 mg/dL; direct bilirubin, 0.7 mg/dL;
reduced as are hemoglobin and hematocrit values, so lactate dehydrogenase, 1902 U/L; prothrombin
that even modest volumes of transfused RBCs will time, 11.8 seconds (normal 11–13 seconds); and
result in hypervolemia. Not only should the total partial thromboplastin time 20.8 seconds (normal,
volume that is transfused be kept modest but RBCs 25–35 seconds). The urine was grossly red but with
should also be administered slowly, the total volume no intact RBCs. The peripheral smear demonstrated
transfused not exceeding 1 mL/kg/hour. One must spherocytes, polychromasia, and nucleated RBCs.
look for evidence of congestive heart failure during The DAT was positive using polyspecific AGS, anti-
and following the transfusion—particularly elevated IgG, and anti-C3. “Least incompatible” (w+ to 2+)
venous pressure and the presence of rales on auscul- units were selected for transfusion using differen-
tation of the chest. Cardiac failure after the adminis- tially adsorbed sera for cross-matching. Before trans-
tration of as little as 200 mL of RBCs can develop up fusion, immune complexes were detected in the
to 6 to 12 hours later and can be fatal. Although serum by several techniques.
diuretics are of value and probably should be given to The patient underwent aggressive transfusion,
patients with diminished cardiac reserve, responses receiving 8 units of packed RBCs over the course of 24
vary, and their administration must not replace close hours, as shown in Figure 10-2. The patient was trans-
clinical observation of the patient. fused to a hematocrit level of greater than 25%. Signs of
DIC developed, however, with the platelet count
POST-TRANSFUSION HEMOGLOBINEMIA, demonstrating a progressive decline and the prothrom-
HEMOGLOBINURIA, AND DISSEMINATED bin time and partial thromboplastin times showing an
INTRAVASCULAR COAGULATION upward trend within 24 hours. One-half hour before the
patient’s death, the following laboratory results were
An additional danger for patients with AIHA relates obtained: prothrombin time, 16.8 seconds; partial
to the fact that the kinetics of RBC destruction always thromboplastin time, 81 seconds; serum fibrinogen, 130
describe an exponential curve of decay, indicating that mg/dL (normal, 150–400 mg/dL); thrombin time, 45
the number of cells removed in a unit of time is a per- seconds (control, 16.5 seconds); fibrin degradation
centage of the number of cells present at the start of products titer, greater than 40 mg/mL; and D-dimer,
this time interval.37 Thus, the more cells present at greater than 1 μg/mL. A diagnosis of DIC was made.
zero time, the greater the absolute number of RBCs The patient became hypotensive shortly thereafter and
that will be destroyed in the unit time span. Indeed, could not be resuscitated.
Blood Transfusion in Autoimmune Hemolytic Anemias 393

Transferred to
UCHC
Steroids
FFP Tx
Platelet Tx
PRBC Tx
IVIG
FIGURE 10-2. Clinical course of AIHA and
DIC in a patient with HIV infection PT, pro- 20 100

PTT (seconds)
thrombin time; PTT, partial thromboplastin

PT (seconds)
time; PRBC, packed red blood cell;
FFP, fresh-frozen plasma; Tx, transfusion;
UCHC, University of Connecticut Health 15 80
Center. (From Bilgrami S, Cable R, Pisciotto
P, Rowland F, Greenberg B: Fatal dissemi-
nated intravascular coagulation and
10 20
pulmonary thrombosis following blood
transfusion in a patient with severe
autoimmune hemolytic anemia and human 200 30
immunodeficiency virus infection.

Hematocrit (%)
Platelet count

Transfusion 1994;34:248–252.) 160


(×109/L)

20
120
80
10
40
0 0
0 24 48 72 96 120
Hours

The autopsy demonstrated widespread multiple required 1 to 2 units of packed RBCs daily to maintain her
pulmonary thrombi. There was widespread thrombo- hemoglobin level at 4.5 to 6.0 g/dL. She experienced
sis of small arterioles of both lungs. Some of the increasingly severe hemoglobinemia and hemoglobinuria
thrombi had begun to organize and others were very associated with transfusions, clearing within a few hours.
acute, a presentation most consistent with the forma- No alloantibodies were demonstrable. On the 10th hos-
tion of thrombi in situ rather than secondary to throm- pital day, a splenectomy was performed without benefit.
boembolic disease. The extensive pulmonary thrombi On the 12th hospital day, several hours following a trans-
resulted in acute right ventricular failure, which was fusion, she developed acute respiratory distress followed
the immediate cause of death. The authors pointed by cardiopulmonary arrest, which did not respond to
out that less aggressive transfusion therapy would resuscitative measures. Autopsy revealed a single large
have sufficed to meet the patient’s tissue oxygen para-aortic lymph node diagnosed as plasmacytoid
needs and concluded that the most likely etiology of malignant lymphoma; death was attributed to multiple
the patient’s DIC was the aggressive transfusion fresh small pulmonary emboli.
therapy. They concluded that their case illustrates the
need to consider the possibility of serious transfusion
complications for patients with AIHA (even when P A T I E N T 2 : A 56-year-old white female, never previ-
alloantibodies have been excluded) and underscores ously pregnant or transfused, was semistuporous at the
the value of the judicious use of transfusion for time of admission to the hospital. She had a hemoglobin
specific indications in this group of patients.5 level of 1.6 g/dL; hematocrit level 6.2%; reticulocytes
Additional examples of unfavorable outcomes of 19%; DAT strongly positive for C3d, moderately positive
transfusion are indicated in the following two case for IgG, and weakly positive for IgM; and all crossmatches
summaries (Patient 1, Patient 2), which were supplied “incompatible.” She was given corticosteroids and 3 units
by Dr. Hugh Chaplin, Jr (Chaplin H Jr, personal of packed group O, Rh-negative RBCs and developed
communication). fever and striking hemoglobinemia and hemoglobinuria,
which improved over the ensuing 3 hours. Her state of
P A T I E N T 1 : A 34-year-old white gravida-1, para-1, consciousness improved, and the hemoglobin level rose
with relapsing primary autoimmune hemolytic anemia for transiently to 5.7 g/dL but was declining rapidly when
4 months, was admitted to the hospital with a hemoglo- she developed severe respiratory distress followed by car-
bin level of 5.0 g/dL; reticulocytes of 16%; DAT strongly diopulmonary arrest, which did not respond to resuscita-
positive for C3d, moderately positive for IgM, and nega- tion. At autopsy, no underlying cause for the AIHA was
tive for IgG; and all crossmatches deemed “incompati- found; death was attributed to multiple fresh small pul-
ble.” Despite high-dose therapy with corticosteroids, she monary emboli.
394 Immune Hemolytic Anemias

Saif and coworkers117 reported a complication of attributable to the transfusion.123 Also, Mollison124
transfusion in a patient with HIV-associated AIHA. points out that the survival of a small volume of
Following transfusion of 1 unit of RBCs, the patient incompatible RBCs might be different from the sur-
developed a pulmonary embolism. The authors cited vival of a larger volume of the same RBCs. Data are
two other reports of thromboembolism in patients not available to determine the rate of destruction of a
with HIV-associated AIHA118,119 and also cited the test dose of RBCs that would predict an acute symp-
case of Bilgrami and associates (reviewed previously), tomatic hemolytic reaction.
although the latter case described DIC rather than Also, the results of an in vivo compatibility test
thromboembolism. could impart a false sense of security, as only a small
These complications of transfusion patients with percentage of transfused RBCs might survive in the 16
AIHA are not common, as emphasized by Salama and to 24 hours following acceptable results in an in vivo
associates,27 who found no instances of a definite survival study.120,125
increase in signs of hemolysis among 53 patients with For example, Silvergleid and coworkers120 des-
AIHA who received blood transfusions. Nevertheless, cribed patients with WAIHA who had adequate
for patients with AIHA, RBCs must be transfused cau- 1-hour RBC survival of a test dose of RBCs and who
tiously and for appropriate indications.5 did not evidence an acute transfusion reaction, but
who had short survival of the transfused RBCs in
subsequent hours. One such patient had a pretrans-
IN VIVO COMPATIBILITY TESTING fusion hematocrit level of 10.5%, a post-transfusion
hematocrit level of 20.6%, but a hematocrit level of
In vivo compatibility testing has been advocated by only 10% just 16 hours after transfusion. Another
some and is based on testing the survival of an aliquot patient with PCH had a 1-hour survival of Tj (a+)
of RBCs from a unit that has been selected for transfu- RBCs of 87%, but only 53% of the radiolabeled cells
sion.56,120 Although such tests are potentially of value survived to 48 hours after transfusion. Still another
in determining the clinical significance of some patient with an alloantibody (anti-Jkb) had 94% sur-
alloantibodies in patients without AIHA, we feel that vival of tagged cells at 1 hour but only 10% survival
such studies are of limited value for patients with at 24 hours after transfusion. Similar data were
AIHA and should never be considered a substitute for reported by Peters and associates,125 who described a
meticulous serologic evaluation and detailed compat- patient with anti-Lub; survival of radiolabeled
ibility testing. This is particularly true because the Lu(b+) RBCs was 84.4% at 1 hour but only 48% at
original use of in vivo survival studies in patients 5 hours and 8.5% at 24 hours after transfusion.
with AIHA occurred at a time when less effective At the present time, in vivo compatibility tests
methods were available for the detection of RBC using radiolabeled RBC seem to be rarely, if ever, per-
alloantibodies in such patients. Using the modern formed in clinical services on patients with AIHA.
compatibility tests described previously, little is to be An alternative method for determining in vivo
gained from in vivo survival studies. survival of transfused RBCs in a patient with AIHA
Some investigators have measured the survival of is to infuse over a period of 20–30 minutes an aliquot
an aliquot of RBCS using 51Cr-tagged cells.120,121 In containing 20–30 mL of RBCs from the unit to be
this procedure, 0.5 to 1 mL of RBCs from a donor unit transfused. The patient is observed for symptoms of
are labeled with 51Cr. If the amount of radioactivity in a hemolytic transfusion reaction, and a blood sample
the plasma, both at 10 and 60 minutes, does not is obtained after the infusion of the “test dose” to
exceed 5% of the radioactivity injected, and if RBC look for hemoglobinemia (and urine, if available, is
survival at 60 minutes is not less than 70%, the expec- observed for hemoglobinuria). The intravascular
tation was that donor RBCs could be transfused with lysis of as little as 5 mL of RBCs will raise the plasma
minimum hazard.71,121 The potential value of this hemoglobin concentration of an adult by about
procedure according to available data is that an 50 mg/100 mL, an amount easily detected by visual
acceptable result in an in vivo compatibility test seems inspection.113 One merely needs to obtain an antico-
to preclude the possibility of an immediate sympto- agulated specimen of blood, centrifuge it, and visu-
matic hemolytic reaction when cells from the whole ally compare it to the plasma obtained prior to the
unit subsequently are transfused.120-122 If the in vivo infusion. Absence of hemoglobinemia and hemoglo-
test is performed after detailed compatibility testing binuria suggests that an acute hemolytic transfusion
using modern techniques, however, and if it demon- reaction will not occur with infusion of the entire
strates short survival of the test cells, it is difficult to unit of blood.
understand what should be done next to select a unit One benefit of this technique is that the test requires
of RBC different from the one used for the in vivo test. evaluation of the patient’s status during the early
Indeed, Meyer and colleagues123 presented data on phase of a transfusion. Close observation of the
11 patients with AIHA who had an in vivo survival of patient is critical, although it obviously should be
51Cr-tagged donor red blood cells of less than 70% in carried out in any case. As a modification of this pro-
1 hour but who nevertheless required transfusion. cedure, one can merely transfuse slowly until the
Only one of these patients had a complication directly equivalent of about 20–30 mL of RBCs have been
Blood Transfusion in Autoimmune Hemolytic Anemias 395

infused, and then obtain a blood sample for visual is maintained at a warm temperature. Johnsen and
inspection of the plasma. One need not stop the trans- coworkers107 reported the results of transfusion of
fusion if the patient remains asymptomatic. 150 mL of prewarmed, packed RBCs (P-positive) to an
An acute hemolytic transfusion serious enough to 18-month-old boy with PCH. The transfusion was fol-
cause immediate hemoglobinemia is not likely to lowed by a temperature rise and passage of red-colored
occur except as a result of ABO incompatibility. Thus, urine. The hemoglobin improved from 4.7 g/dL to
little information, if any, is provided about the pres- 12.4 g/dL and remained at that level.
ence of other alloantibodies that have the potential to In the absence of extensive data, logic must prevail.
cause a hemolytic transfusion reaction. Even if no Our experience in CAS has been consistent with
immediate adverse effects are noted, the survival of Dacie’s view, although in some instances we have
the full unit is not likely to be better than that of the empirically used an in-line blood warmer for seriously
patient’s own RBCs. ill patients. The use of an in-line blood warmer would
More important, if a careful serologic study has been appear indicated if the patient has either severe PCH or
done using the techniques and principles already florid CAS. It is also logical to keep the patient warm,
described to select the best possible blood for transfu- even if the efficacy of such a maneuver has not been
sion, and if an in vivo test nevertheless demonstrates a proven.
very short RBC survival, there is no logical way to If blood is to be warmed, it must be done properly.
select an alternative unit. Certainly, a patient should Unmonitored or uncontrolled heating of blood is
never be denied a needed transfusion because labora- extremely dangerous and should not be attempted.
tory data (whether from a compatibility test or from an RBCs heated too much are rapidly destroyed in vivo
RBC survival study) suggest that the transfused blood and can be lethal.37 Very efficient in-line blood
will not survive well. If the patient requires transfusion, warmers are available that are simple, efficient, and
such incompatible blood must still be given in the hope safe to use. Guidelines for the use of blood warming
of obtaining temporary benefit. devices have been published.127
In conclusion, we feel that the significance of in vivo
survival studies with respect to transfusion of patients
with AIHA has been overemphasized. THE USE OF RBC SUBSTITUTES

Considerable progress has been made in the develop-


THE USE OF WARM BLOOD FOR ment of RBC substitutes, and the availability of O2-
PATIENTS WITH COLD AGGLUTININ carrying therapeutic agents for clinical use could
SYNDROME AND PAROXYSMAL affect the practice of transfusion medicine.25,26,128-132
Mullon and associates26 described the use of a poly-
COLD HEMOGLOBINURIA merized bovine hemoglobin in a patient with severe
AIHA. The patient had life-threatening AIHA that
Eminent authorities offer sharply differing opinions was refractory to treatment with high doses of gluco-
concerning the need for warm blood when transfu- corticoids, plasmapheresis, splenectomy, and intra-
sing patients with CAS. Dacie33 states that properly venous immune globulin. To maintain a hematocrit
crossmatched blood can probably be transfused with greater than 12% and a hemoglobin level greater than
safety if run in at a slow drip rate, in which case there 4.0 g/dL, as many as 8 units of RBC per day were
is probably no need to attempt to warm it above needed; she received a total of 53 units of RBCs before
room temperature. By contrast, Rosenfield and initiation of therapy with the hemoglobin solution.
Jagathambol37 state that patients with cold agglutinin During RBC transfusions, the patient developed
disease must receive warmed blood. Mollison and symptoms attributed to acute hemolysis, including
colleagues71 state that blood should be warmed before fever, nausea, and back pain. Ischemic changes were
transfusion to patients with cold agglutinin syn- present in the electrocardiogram when the hemoglo-
drome, and they also recommend that the patient be bin was at a level of about 4.0 g/dL.
kept warm. Wallace126 comments that hemolytic reac- A total of 11 units of a sterile solution of gluteralde-
tions are unlikely, provided that the donor blood is hyde-polymerized bovine hemoglobin were trans-
warmed to body temperature and the recipient is kept fused over a 7-day period. Five units were
warm. As is evident, the question has not been administered in response to clinical evidence of
studied in much depth, and no RBC survival studies ischemia, three as part of volume resuscitation during
are available comparing survival of blood transfused an episode of septic shock, and three to maintain the
at various temperatures. total hemoglobin level above 4 g/dL. This therapy
With regard to PCH, Rausen22 does report that even resulted in relief of ischemia, and no clinically impor-
compatible Tj(a-) blood needs to be warmed to 37°C tant adverse hemodynamic responses occurred. She
before transfusion. Wallace126 states that transfusion of received further treatment for her AIHA with
RBCs of the common P groups is unlikely to precipitate cyclophosphamide and developed profound neutro-
an acute hemolytic transfusion reaction in PCH pro- penia, gram-negative septic shock, and gram-negative
vided that the blood is warmed to 37°C and the patient pelvic osteomyelitis. Subsequent immunosuppressive
396 Immune Hemolytic Anemias

therapy with cyclosporine led to sustained improve- technical procedures, and there could be a longer
ment in her AIHA, transfusions were no longer neces- delay in selecting an alternative unit.
sary, and 8 months after discharge the patient was
well, with a hematocrit consistently greater than 35%.
There are indications that cell-free hemoglobin sub- USE OF WASHED RBCs
stances might support bacterial virulence by offering
a ready supply of iron, thus sustaining bacterial repli- Previously, washed RBCs have been used to reduce
cation and inhibiting neutrophil function.133,134 In the the incidence of febrile nonhemolytic transfusion
case of this patient, however, it is unclear what role reactions for the same reason indicated for the use of
the hemoglobin solution might have had in promot- leukocyte-reduced RBCs. Washed RBCs have also
ing infectious complications, given her neutropenia been suggested as possibly being of benefit to
and preexisting AIHA. patients with AIHA because they are free of comple-
Raff and colleagues128 reported a 40-year-old allo- ment components that will be in any plasma-
immunized woman with sickle cell anemia who under- containing blood product. The data supporting this
went hip replacement surgery. Periopertively, she justification for the washed RBCs is inconclusive,
received 4 units of matched frozen RBCs. Seven days however, and we do not recommend washed RBCs
postoperatively, however, she was readmitted with based on this rationale.
generalized pain, fever, dyspnea, and positive blood The recommendation for the use of washed RBCs
and urine cultures. Hemoglobin decreased to for transfusion of patients with AIHA is largely
2.8 g/dL, with no improvement after transfusion of 5 based on the data of Evans and coworkers,35,135 who
matched units, findings suggestive of the sickle cell reported their experience using packed RBCs or
hemolytic transfusion reaction syndrome (see Chapter washed RBCs in transfusing one patient with CAS.
14). She was treated with antibiotics, IVIG, corticos- The cold antibody agglutinated RBCs to a titer of
teroids, erythropoietin, and iron. In addition, she 500,000 at 4°C and caused lysis of normal RBCs both
received five daily 1-unit transfusions of PolyHeme, a at room temperature and at 37°C (maximal at room
polymerized human hemoglobin. Pain and dyspnea temperature). A transfusion of 2 units of packed
improved markedly, and she was discharged 10 days RBCs was given without subjective reaction,
after the start of treatment with a hemoglobin level of although hemoglobinuria occurred following com-
7.8 g/dL. The authors commented that the multiple pletion of the transfusion. A subsequent transfusion
therapeutic interventions make it difficult to assess the of 3 units of washed RBCs was given without rise in
contribution of the hemoglobin solution. Nevertheless, plasma hemoglobin. The patient’s serum comple-
they suggested that hemoglobin-based oxygen carriers ment was “always low.” Although the data are far
could be a valuable contribution to treatment “in these from conclusive, the authors speculated that the
cases that are every blood banker’s worst nightmare.” small amount of plasma present in the first transfu-
Other reviews have been published of the use of sion of RBCs provided the necessary complement for
artificial oxygen carriers, primarily in the setting of the hemolysis that followed.
acute blood loss.129-132 Such reports are generally Further studies by Evans and associates35 indicated
enthusiastic, although Tremper,131 whose editorial that transfusion of large volumes of RBCs to patients
regarding perfluorochemical compounds bears the with CAS caused a lowering of serum complement.
subtitle, “The continued search for an indication,” 51Cr-labeled normal RBCs survived longer after RBC
submits a note of caution. Nevetheless, he remains transfusions than before, and the authors suggested
hopeful that the broader utility of such compounds that the reduction in serum complement levels might
might be proven. have been responsible for the improved survival.
The phenomenon reported by Evans requires more
data for documentation. Serum complement is ordi-
USE OF LEUKOCYTE-REDUCED RBCs narily not reduced in patients with warm- or cold-
antibody AIHA, and we would not recommend the
Some investigators recommend using leukocyte- use of washed RBCs based on this rationale because of
reduced RBCs for patients with AIHA to avoid a the limited information available.
febrile nonhemolytic transfusion reaction, which
would confound an evaluation of a possible hemolytic AUTOLOGOUS BLOOD
reaction. The interpretation of a febrile reaction in a
seriously ill patient with AlHA is difficult and could TRANSFUSION IN AIHA
lead to unnecessary cessation or delay of transfusion.
These reactions could be reduced in incidence by the Three brief reports have suggested the use of autolo-
use of leukocyte-reduced RBCs. Such a situation pro- gous RBCs for transfusion of patients with AIHA.
vides a justification for their use in AIHA, as the com- Goldfinger and colleagues136 demonstrated that auto-
patibility test procedures could be labor intensive and logous frozen-stored RBCs could be practical for some
time consuming. Thus, unnecessary cessation of the patients with AIHA. In one patient with IgM-
transfusion is more costly because of the additional mediated AIHA, normal in vivo recovery was demon-
Blood Transfusion in Autoimmune Hemolytic Anemias 397

strated, although in vitro RBC loss was 40%. In 12. Hauke G, Fauser AA, Weber S, Maas D: Reticulocytopenia in
patients who had positive DATs but not hemolytic severe autoimmune hemolytic anemia (AIHA) of the warm
anemia, in vitro RBC loss was only 2% to 14%, and in antibody type. Blut 1983;46:321–327.
13. Carapella de Luco E, Stegagno M, Morino G, Ballati G,
vivo recovery and survival were normal. Storing Lorenzini F: Prolonged reticulocytopenia and transient ery-
RBCs in the frozen state may be considered for throid hypoplasia in a child with autoimmune haemolytic
patients who go into remission while on therapy for anaemia. Haematologica 1984;69:315–320.
AIHA. In the event of later relapse, the RBCs may be 14. Greenberg J, Curtis-Cohen M, Gill FM, Cohen A: Prolonged
reticulocytopenia in autoimmune hemolytic anemia of child-
transfused without fear of alloantibody-induced hood. J Pediatr 1980;97:784–786.
hemolysis. Because of autoantibody in the patient’s 15. Cazzola M, Barosi G, Ascari E: Cold haemagglutinin disease
serum, however, RBC survival is not likely to be better with severe anaemia, reticulocytopenia and erythroid bone
than that of an allogeneic unit. marrow. Scand J Haematol 1983;30:25–29.
Parr and Ballard137 reported on a patient with 16. Brown KE: Haematological consequences of parvovirus B19
infection. Baillieres Best Pract Res Clin Haematol
methyldopa-induced positive DAT and IAT who 2000;13:245–259.
required surgery. The patient had equivocal evidence 17. Bertrand Y, Lefrere JJ, Leverger G, et al: Autoimmune hae-
of hemolysis, consisting of an elevated LDH with a molytic anaemia revealed by human parvovirus linked
normal hemoglobin and hematocrit. The patient’s erythroblastopenia. Lancet 1985;ii:382–383.
18. Chitnavis VN, Patou G, Makar YF, Kendra JR: B19 parvovirus
serum antibody reacted with all normal RBCs tested. induced red cell aplasia complicating acute cold antibody
Rather than wait for a fall in the autoantibody titer fol- mediated haemolytic anaemia. Br J Haematol 1990;76:
lowing cessation of the drug, 3 units of autologous 433–434.
blood were stored in the liquid state and used during 19. Mangan KF, Besa EC, Shadduck RK, Tedrow H, Ray PK:
surgery. A similar approach was used by Snyder and Demonstration of two distinct antibodies in autoimmune
hemolytic anemia with reticulocytopenia and red cell aplasia.
Spivack138 for two patients. Although evidence indi- Exp Hematol 1984;12:788–793.
cates that drug-induced autoantibodies will not cause 20. Meyer RJ, Hoffman R, Zanjani ED: Autoimmune hemolytic
shortened survival of homologous blood if the anemia and periodic pure red cell aplasia in systemic lupus
patient’s own RBC survival is normal, the use of erythematosus. Am J Med 1978;65:342–345.
21. Todd D: Diagnosis of haemolytic states. Clinics Haematol
autologous blood ensures that no alloantibody- 1975;4:63–81.
induced transfusion reaction will occur. In instances 22. Rausen AR, LeVine R, Hsu TC, Rosenfield RE: Compatible
in which it is inconvenient or impossible to delay transfusion therapy for paroxysmal cold hemoglobinuria.
surgery until an adequate number of autologous units Pediatrics 1975;55:275–278.
are collected, the warm autoadsorption technique or 23. Swisher SN, Petz LD: Transfusion therapy for chronic anemic
states. In Petz LD, Swisher SN, Kleinman S, Spence RK,
other serologic methods as described previously for Strauss RG (eds): Clinical Practice of Transfusion Medicine,
patients with warm-antibody AIHA may be used to 3rd ed. New York: Churchill Livingstone, 1996:451–467.
exclude the presence of alloantibody. 24. Hillman RS: Blood-loss anemia. Postgrad Med 1978;64:88–94.
25. Klein HG: The prospects for red-cell substitutes. N Engl J Med
2000;342:1666–1668.
26. Mullon J, Giacoppe G, Clagett C, McCune D, Dillard T:
R E F E R E N C E S Transfusions of polymerized bovine hemoglobin in a patient
with severe autoimmune hemolytic anemia. N Engl J Med
1. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias, 2000;342:1638–1643.
1st ed. New York: Churchill Livingstone, 1980. 27. Salama A, Berghofer H, Mueller-Eckhardt C: Red blood cell
2. Petz LD: Red cell transfusion problems in immunohemato- transfusion in warm-type autoimmune haemolytic anaemia.
logic disease. Annu Rev Med 1982;33:355–361. Lancet 1992;340:1515–1517.
3. Petz LD: Blood transfusion in hemolytic anemias. Immuno- 28. Sokol RJ, Hewitt S, Booker DJ, Morris BM: Patients with red
hematology 1999;15:15–23. cell autoantibodies: Selection of blood for transfusion. Clin
4. Petz LD: Blood transfusion in acquired hemolytic anemia. In Lab Haematol 1988;10:257–264.
Petz LD, Swisher SN, Kleinman S, Spence RK, Strauss RG 29. Habibi B, Homberg JC, Schaison G, Salmon C: Autoimmune
(eds): Clinical Practice of Transfusion Medicine, 3rd ed. New hemolytic anemia in children: A review of 80 cases. Am J Med
York: Churchill Livingstone, 1996:469–499. 1974;56:61–69.
5. Garratty G, Petz LD: Transfusing patients with autoimmune 30. Pirofsky B: Autoimmunization and the Autoimmune
haemolytic anaemia. Lancet 1993;341:1220. Hemolytic Anemias. Baltimore: Williams & Wilkins, 1969.
6. Petz LD: Transfusing the patient with autoimmune hemolytic 31. Pirofsky B, Bardana EJ Jr: Autoimmune hemolytic anemia. II.
anemia. Clin Lab Med 1982;2:193–210. Therapeutic aspects. Ser Haematol 1974;7:376–385.
7. Jefferies LC: Transfusion therapy in autoimmune hemolytic 32. Audet AM, Popovsky MA, Andrzejewski C: Current transfu-
anemia. Hematol Oncol Clin North Am 1994;8:1087–1104. sion practices in orthopedic surgery patients: A multi-
8. Conley CL, Lippman SM, Ness P: Autoimmune hemolytic institutional study [abstract]. Blood 1995;86(Suppl.):853a.
anemia with reticulocytopenia: A medical emergency. JAMA 33. Dacie JV: Autoimmune Haemolytic Anaemias (AIHA):
1980;244:1688–1690. Treatment. In The Haemolytic Anaemias, 3rd ed. vol. 3, The
9. Conley CL, Lippman SM, Ness PM, Petz LD, Branch DR, Auto-Immune Haemolytic Anaemias. New York: Churchill
Gallagher MT: Autoimmune hemolytic anemia with reticulo- Livingstone, 1992:452–520.
cytopenia and erythroid marrow. N Engl J Med 1982; 34. Masouredis SP, Chaplin H: Transfusion Management of
306:281–286. autoimmune hemolytic anemia. In Chaplin H (ed): Immune
10. Crosby WH, Rappaport H: Reticulocytopenia in autoimmune Hemolytic Anemias. New York: Churchill Livingstone,
hemolytic anemia. Blood 1956;11:929–936. 1985:177–205.
11. Liesveld JL, Rowe JM, Lichtman MA: Variability of the ery- 35. Evans RS, Turner E, Bingham M, Woods R: Chronic hemolytic
thropoietic response in autoimmune hemolytic anemia: anemia due to cold agglutinins. II. The role of C’ in red cell
Analysis of 109 cases. Blood 1987;69:820–826. destruction. J Clin Invest 1968;47:691–701.
398 Immune Hemolytic Anemias

36. Haemolytic Transfusion Reactions. In Mollison PL, Engelfriet 60. Champagne K, Moulds MK: Autoadsorptions for the detec-
CP, Contreras M (eds): Blood Transfusion in Clinical tion of alloantibodies—Should polyethylene glycol be used?
Medicine, 10th ed. Oxford: Blackwell Science, 1997:358–389. Transfusion 1996;36:384.
37. Rosenfield RE, Jagathambal: Transfusion therapy for autoim- 61. Miller R, Unger P, Shapiro A: Rapid adsorption of warm
mune hemolytic anemia. Semin Hematol 1976;13:311–321. autoantibodies using polyethylene glycol [abstract]. Trans-
38. Edwards JM, Moulds JJ, Judd WJ: Chloroquine dissociation of fusion 1996;36(Suppl.):20S.
antigen-antibody complexes: A new technique for typing red 62. Hernandez-Jorda M, Zamora C, Duarte R, et al: Polyethylene
blood cells with a positive direct antiglobulin test. Transfusion glycol (PeG) in microadsorption procredures to detect red cell
1982;22:59–61. alloantibodies in the presence of autoantibodies. Transfus
39. Branch DR, Hian AL, Carlson F, Maslow WC, Petz LD: Med 1998;8:278.
Erythrocyte age-fractionation using a Percoll-Renografin 63. Barron CL, Brown MB: The use of polyethylene glycol (PEG)
density gradient: Application to autologous red cell antigen to enhance the adsorption of autoantibodies. Immumno-
determinations in recently transfused patients. Am J Clin hematology 1997;13:119–122.
Pathol 1983;80:453–458. 64. Judd WJ, Dake L: PEG adsorption of autoantibodies csause
40. Brecher M (ed): Technical Manual, 14th ed. Bethesda, MD: loss of concomitant allo-antibody. Immunohematology
American Association of Blood Banks, 2002. 2002;17:82–85.
41. Reid ME, Toy PT: Simplified method for recovery of autolo- 65. Overbeeke MAM, Porcelijn L: The detection of alloantibodies
gous red blood cells from transfused patients. Am J Clin against red cells in patients with warm-type autoimmune
Pathol 1983;79:364–366. haemolytic anaemia. International Forum. Vox Sang 2000;78:
42. Vengelen-Tyler V, Gonzales B: Reticulocyte rich RBCs will 203–204.
give weak reactions with many blood typing antisera 66. Combs MR, Eveland D, Jewett-Keefe B, Telen MJ: The use of
[abstract]. Transfusion 1985;25:476. Polyethylene Glycol in adsorptions: More evidence that anti-
43. Wenk RE, Chiafari PA: DNA typing of recipient blood after bodies may be missed. Transfusion 2001;41:30S.
massive transfusion. 1997;37:1108–1110. 67. Tocci LJ, Reeves CT: Cost saving method for the preparation
44. Reid ME, Rios M, Powell VI, Charles-Pierre D, Malavade V: of differential adsorbing cells [abstract]. Transfusion 1998;
DNA from blood samples can be used to genotype patients 38(Suppl.):89S.
who have recently received a transfusion. Transfusion 68. Petz LD, Branch DR: Serological tests for the diagnosis of
2000;40:48–53. immune hemolytic anemias. In McMillan R (ed): Methods in
45. Castilho L, Rios M, Pellegrino J, Rodrigues A, Costa FF: Blood Haematology: Immune Cytopenias. New York: Churchill
group genotyping for the management of patients with Livingstone, 1983:9–48.
“warm” antibody-induced hemolytic anemia (WAIHA) 69. Petz LD, Garratty G: Blood Transfusion in Autoimmune
[abstract]. Blood 2001;98:62a. Hemolytic Anemias. In Petz LD, Garratty G (eds): Acquired
46. Branch DR, Petz LD: Detecting alloantibodies in patients with Immune Hemolytic Anemias, 1st ed. New York: Churchill
autoantibodies. Transfusion 1999;39:6–10. Livingstone, 1980:358–391.
47. Morel PA, Bergen MO, Frank BA: A simple method for the 70. Laine EP, Leger RM, Arndt PA, Calhoun L, Garratty G, Petz
detection of allo-antibody in the presence of warm autoanti- LD: In vitro studies of the impact of transfusion on the detec-
body [abstract]. Transfusion 1978;18:388. tion of alloantibodies after autoadsorption. Transfusion
48. Branch DR, Petz LD: A new reagent ZZAP having multiple 2000;40:1384–1387.
applications in immunohematology. Am J Clin Pathol 71. Mollison PL, Engelfriet CP, Contreras M (eds): Blood Transfusion
1982;78:161–167. in Clinical Medicine, 10th ed. Oxford: Blackwell Science, 1997.
49. Wallhermfechtel MA, Pohl BA, Chaplin H: Alloimmunization 72. Reid ME, Toy PT: Use of preserved autologous red blood cells
in patients with warm autoantibodies: A retrospective study to absorb warm autoantibodies from the serum of patients
employing three donor alloabsorptions to aid in antibody receiving blood transfusion therapy. Vox Sang 1984;46:355–359.
detection. Transfusion 1984;24:482–485. 73. Garratty G: Autoimmune hemolytic anemia. In Garratty G
50. Laine ML, Beattie KM: Frequency of alloantibodies accompa- (ed): Immunobiology of Transfusion Medicine. New York:
nying autoantibodies. Transfusion 1985;25:545–546. Marcel Dekker, 1994:493–521.
51. James P, Rowe GP, Tozzo GG: Elucidation of alloantibodies in 74. Garratty G: Target antigens for red cell-bound autoantibodies.
autoimmune haemolytic anaemia. Vox Sang 1988;54:167–171. In Nance SJ (ed): Clinical and serological aspects of immuno-
52. Issitt PD, Combs MR, Bumgarner DJ, Allen J, Kirkland A, hematology. Arlington, VA: American Association of Blood
Melroy-Carawan H: Studies of antibodies in the sera of Banks, 1991:33.
patients who have made red cell autoantibodies. Transfusion 75. Mollison PL: Measurement of survival and destruction of red
1996;36:481–486. cells in haemolytic syndromes. Br Med Bull 1959;15:59.
53. Leger RM, Garratty G: Evaluation of methods for detecting 76. Red cell antibodies against self antigens, bound antigens and
alloantibodies underlying warm autoantibodies. Transfusion induced antigens. In Mollison PL, Engelfriet CP, Contreras M
1999;39:11–16. (eds): Blood Transfusion in Clinical Medicine, 10th ed. Oxford:
54. So CC, Wong KF, Yu PH, Kwan AM, Lee AW: Alloimmunization Blackwell Science, 1997:213–240.
in Chinese with warm autoimmune haemolytic anaemia– 77. Salmon C: Autoimmune hemolytic anemia. In Bach J,
Incidence and characteristics. Transfus Med 2000;10:141–143. Swenson RS, Schwartz RS (eds): Immunology. New York:
55. Oyen R, Angeles ML: A simple screening method to evaluate Wiley & Sons, 2001:675.
the presence of alloantibodies with concomitant warm 78. Borne AE von dem, Engelfriet CP, Beckers D, Van Loghem JJ:
autoantibodies. Immunohem 1995;11:85–87. Autoimmune haemolytic anaemias. IV. Biochemical studies of
56. Dorner IM, Parker CW, Chaplin H Jr: Autoagglutination red cells from patients with autoimmune haemolytic anaemia
developing in a patient with acute renal failure: Character- with incomplete warm autoantibodies. Clin Exp Immunol
ization of the autoagglutinin and its relation to transfusion 1971;8:377–388.
therapy. Br J Haematol 1968;14:383–394. 79. Hollander L: Study of the erythrocyte survival time in a case
57. Liew YW, Duncan N: Polyethylene glycol in autoadsorption of of acquired haemolytic anaemia. Vox Sang 1954;4:164.
serum for detection of alloantibodies. Transfusion 1995;35:713. 80. Crowley LV, Bouroncle BA: Studies on the specificity of autoan-
58. Liew YW, Duncan N: Autoadsorptions for the detection of tibodies in acquired hemolytic anemia. Blood 1956;11:700.
alloantibodies—Should polyethylene glycol be used? Trans- 81. Wiener AS, Gordon EB, Russow E: Observations on the nature
fusion 1996;36:384. of the autoantibodies in a case of acquired hemolytic anemia.
59. Cheng CK, Wong ML, Lee AW: PEG adsorption of autoanti- Ann Int Med 1957;47:1.
bodies and detection of alloantibodies in warm autoimmune 82. Ley AM, Mayer K, Harris JP: Observations on a “specific autoan-
hemolytic anemia. Transfusion 2001;41:13–17. tibody.” Proc 6th Congr Int Soc Blood Trans, Boston, 1958:148.
Blood Transfusion in Autoimmune Hemolytic Anemias 399

83. Hogman C, Killander J, Sjolin S: A case of idiopathic autoim- 107. Johnsen HE, Brostrphom K, Madsen M: Paroxysmal cold
mune haemolytic anaemia due to anti-e. Acta Paediat haemoglobinuria in children: 3 cases encountered within a
1960;49:270. period of 7 months. Scand J Haematol 1978;20:413–416.
84. Bell CA, Zwicker H, Sacks HJ: Autoimmune hemolytic 108. O’Neill BJ, Marshall WC: Paroxysmal cold haemoglobinuria
anemia: Routine serologic evaluation in a general hospital and measles. Arch Dis Child 1967;42:183–186.
population. Am J Clin Pathol 1973;60:903–911. 109. Bird GW, Wingham J, Martin AJ, et al: Idiopathic non-
85. Dacie JV. The Haemolytic Anaemias. Part II. Autoimmune syphilitic paroxysmal cold haemoglobinuria in children. J Clin
Haemolytic Anaemias, 2nd ed. London: J. & A Churchill, Pathol 1976;29:215–218.
1962. 110. Wolach B, Heddle N, Barr RD, Zipursky A, Pai KR, Blajchman
86. Petz LD. Transfusing patients with autoimmune hemolytic MA: Transient Donath-Landsteiner haemolytic anaemia. Br J
anemia: How should a clinician deal with “least incompatible” Haematol 1981;48:425–434.
units? Education Manual of the American Society of 111. Sokol RJ, Hewitt S, Stamps BK: Autoimmune haemolysis
Hematology 2002;449–454. associated with Donath-Landsteiner antibodies. Acta
87. Standards for Blood Banks and Transfusion Services, 22nd ed. Haematol 1982;68:268–277.
Bethesda, MD: American Association of Blood Banks, 2003. 112. Lindgren S, Zimmerman S, Gibbs F, Garratty G: An unusual
88. Shulman IA, Nelson JM, Nakayama R: When should antibody Donath-Landsteiner antibody detectable at 37 degrees C by
screening tests be done for recently transfused patients? the antiglobulin test. Transfusion 1985;25:142–144.
Transfusion 1990;30:39–41. 113. Chaplin H Jr: Special problems in transfusion management of
89. Shirey RS, Boyd JS, Parwani AV, Tanz WS, Ness PM, King KE: patients with autoimmune hemolytic anemia. In Bell CA (ed):
Prophylactically antigen-matched donor blood for patients A seminar on Laboratory Management of Hemolysis.
with warm autoantibodies: An algorithm for transfusion Washington, DC: American Association of Blood Banks,
management. Transfusion 2002;42:1435–1441. 1979:135–150.
90. Lau FY, Wong R, Chan NPH, et al: Provision of phenotype- 114. Gurgey A, Yenicesu I, Kanra T, et al: Autoimmune hemolytic
matched blood units: No need for pre-transfusion antibody anemia with warm antibodies in children: Retrospective
screening. Haematologica 2001;86:742–748. analysis of 51 cases. Turk J Pediatr 1999;41:467–471.
91. Garratty G, Petz LD: Approaches to selecting blood for trans- 115. Heidemann SM, Sarniak SA, Sarniak AP: Exchange transfu-
fusion to patients with autoimmune hemolytic anemia. sion for severe autoimmune hemolytic anemia. Amer J Pediatr
Transfusion 2002;42:1390–1392. Hematol Oncol 1987;9:302–304.
92. Shirey RS, Ness PM: New concepts of delayed hemolytic 116. Bilgrami S, Cable R, Pisciotto P, Rowland F, Greenberg B: Fatal
transfusion reactions. In Nance SJ (ed): Clinical and Basic disseminated intravascular coagulation and pulmonary
Science: Aspects of Immunohematology. Arlington, VA: thrombosis following blood transfusion in a patient with
American Association of Blood Banks, 1991:179. severe autoimmune hemolytic anemia and human immuno-
93. Shirey RS, King KE: Alloimmunization to blood group anti- deficiency virus infection. Transfusion 1994;34:248–252.
gens. In Anderson KE, Ness PM (eds): Scientific Basis of 117. Saif MW, Morse EE, Greenberg BR: HIV-associated autoim-
Transfusion Medicine: Implications for Clinical Practice. mune hemolytic anemia complicated by pulmonary
Philadelphia: WB Saunders, 2000:393–400. embolism following a red blood cell transfusion: Case report
94. Garratty G, Petz LD, Hoops JK: The correlation of cold agglu- and review of the literature. Conn Med 1998;62:67–70.
tinin titrations in saline and albumin with haemolytic 118. David A, Thomas M: Upperextremity arterial thrombosis
anaemia. Brit J Haemat 1977;35:587–595. associated with immunohemolytic anemia. Surgery
95. Giblett ER: Blood group alloantibodies: An assessment of 1996;119:354–355.
some laboratory practices. Transfusion 1977;17:299–308. 119. Ries M, Simon S, Hummer HP, Leipold G: Acute mesenteric
96. Stroup M, MacIlroy M: Evaluation of the albumin antiglobu- vein thrombosis: A rare complication after splenectomy due to
lin technique in antibody detection. Transfusion 1965;5:184. autoimmune hemolytic anemia in childhood. Monatssch
97. Dzik WH, Yang R, Blank J: Rabbit erythrocyte stroma treat- Kinderheilkd 1993;141:779–781.
ment of serum interferes with recognition of delayed 120. Silvergleid AJ, Wells RF, Hafleigh EB, et al: Compatibility test
hemolytic transfusion reaction. Transfusion 1986;26:303–304. using chromium-labeled red blood cells in crossmatch posi-
98. Deutsch HF, Morton JI: Dissociation of human serum tive patients. Transfusion 1978;18:8–14.
macroglobulins. Science 1957;125:600. 121. International Committee for Standardization in Haematology:
99. Grubb R, Swahn B: Destruction of some agglutinins but not of Recommended methods for radioisotope red cell survival
others by two sulfhydryl compounds. Acta Path Microbiol studies. Blood 1971;38:378.
Scand 1958;43:305. 122. Mayer K, Bettigole RE, Harris JP, D’Amaro J: Test in vivo to
100. Pirofsky B, Rosner ER: DTT test: A new method to differen- determine donor blood compatibility. Transfusion 1968;8:
tiate IgM and IgG erythrocyte antibodies. Vox Sang 1974; 8–32.
27:480–488. 123. Meyer K, Chin B, Magnes J, Harris JP, Tegoli J: Further experi-
101. Olson PR, Weiblen BJ, O’Leary JJ, Moscowitz AJ, McCullough ences with the in vivo crossmatch and transfusion of Coombs
J: A simple technique for the inactivation of IgM antibodies incompatible red cells [abstract]. XVII Congress of
using dithiothreitol. Vox Sang 1976;30:149–159. International Society of Hematology and XV Congress of
102. Freedman J, Masters CA, Newlands M, Mollison PL: Optimal International Society of Blood Transfusion Book of Abstracts,
conditions for the use of sulphydryl compounds in dissociat- 1978;49.
ing red cell antibodies. Vox Sang 1976;30:231–239. 124. Red cell incompatability in vivo. In Mollison PL, Engelfriet
103. Van Loghem JJ, Peetom F, Van der Hart M, et al: Serological CP, Contreras M (eds): Blood Transfusion in Clinical
and immunochemical studies in hemolytic anemia with high- Medicine, 10th ed. Oxford: Blackwell Science, 1997:315–357.
titer cold agglutinins. Vox Sang 1963;8:33. 125. Peters B, Reid ME, Ellisor SS, Avoy DR: Red cell survival
104. Woll JE, Smith CM, Nusbacher J: Treatment of acute cold studies of Lub incompatible blood in a patient with anti-Lub
agglutinin hemolytic anemia with transfusion of adult i RBCs. [abstract]. Transfusion 1978;18:623.
JAMA 1974;229:1779–1780. 126. Wallace J: Blood Transfusion for Clinicians. New York:
105. Sabio H, Jones D, McKie VC: Biphasic hemolysin hemolytic Churchill Livingstone, 1977.
anemia: Reappraisal of an acute immune hemolytic anemia of 127. AuBuchon JP: Guidelines for the Use of Blood Warming
infancy and childhood. Am J Hematol 1992;39:220–222. Devices. Bethesda, MD: American Association of Blood
106. Nordhagen R, Stensvold K, Winsnes A, Skyberg D, Storen A: Banks, 2001.
Paroxysmal cold haemoglobinuria: The most frequent acute 128. Raff JP, Dobson CE, Tsai HM: Transfusion of polymerised
autoimmune haemolytic anaemia in children? Acta Paediatr human haemoglobin in a patient with severe sickle-cell
Scand 1984;73:258–262. anaemia. Lancet 2002;360:464–465.
400 Immune Hemolytic Anemias

129. Spahn DR: Artificial oxygen carriers: Status 2002. Vox Sang 134. Litwin MS, Walter CW, Ejarque P, Reynolds ES: Synergistic
2002;83 (Suppl. 1):281–285. toxicity of gram-negative bacterial and free colloidal hemo-
130. Spahn DR, Waschke KF, Standl T, et al: Use of perflubron globin. Ann Surg 1963;157:485–493.
emulsion to decrease allogeneic blood transfusion in high- 135. Evans RS, Bingham M, Turner E: Autoimmune hemolytic
blood-loss non-cardiac surgery: Results of a European phase 3 disease: Observations of serologic reactions and disease activ-
study. Anesthesiology 2002;97:1338–1349. ity. Ann NY Acad Sci 1965;124:422.
131. Tremper KK: Perfluorochemical “red blood cell substitutes”: 136. Goldfinger D, Connelly M, Kellum S, Rosenbaum D: Transfu-
The continued search for an indication. Anesthesiology sion of frozen autologous red cells in patients with positive
2002;97:1333–1334. direct antiglobulin tests [abstract]. Blood 1979;54(Suppl. 1):123a.
132. Gould SA, Moore EE, Hoyt DB, et al: The life-sustaining 137. Parr GV, Ballard JO: Autologous blood transfusion with
capacity of human polymerized hemoglobin when red cells methyldopa induced positive direct antiglobulin test. Trans-
might be unavailable. J Am Coll Surg 2002;195:445–452. fusion 1980;20:119.
133. Griffiths E, Cortes A, Gilbert N, Stevenson P, MacDonald S, 138. Snyder EL, Spivack M: Clinical and serologic management of
Pepper D: Haemoglobin-based blood substitutes and sepsis. patients with methyldopa-induced positive antiglobulin tests.
Lancet 1995;345:158–160. Transfusion 1979;19:313–316.
C H A P T E R 1 1

Management of
Autoimmune Hemolytic
Anemias

This chapter summarizes the WARM ANTIBODY AUTOIMMUNE


information available on ther- HEMOLYTIC ANEMIA
apy of autoimmune hemolytic
anemias and suggests a reasonable approach to thera-
peutic management, while recognizing a lack of Corticosteroid Therapy
controlled clinical trials to clarify specific details INITIAL MANAGEMENT
(Table 11-1).
In the 23 years since the publication of the first The one aspect of therapy about which there is
edition of this book, startling advances in scientific uniform agreement is that corticosteroids should be
knowledge have taken place. Indeed, entirely new an initial therapeutic tool in treating patients with
fields have been developed that were not even in autoimmune hemolytic anemia (AIHA) of warm anti-
existence two decades ago. It is disappointing, there- body type. Although detailed studies are not available
fore, that the advances in the management of concerning the efficacy of individual corticosteroids,
immune hemolytic anemias have been modest. there seems to be no documented advantage of any
Indeed, many of the comments about management agent over prednisone or prednisolone. A number of
made in our first edition in 19801 still hold true today, guidelines for the use of corticosteroids in warm anti-
and treatment protocols of yesteryear (or yester- body (WAIHA) have been published, and the follow-
century) are still reiterated in recent medical litera- ing is a composite of these opinions.2-11
ture. Nevertheless, some newer forms of therapy are Prednisone may be used at a dose of 1–1.5 mg/kg/
being introduced. day, equivalent to about 60–100 mg per day for an adult

401
402 Immune Hemolytic Anemias

of average size.9 This therapy should be maintained for


TABLE 11-1. THERAPEUTIC OPTIONS IN 1 to 3 weeks. Data in the older medical literature
AUTOIMMUNE HEMOLYTIC ANEMIAS indicates that the initial response is often excellent, with
about 80% of patients experiencing prompt reduction in
Warm Antibody Autoimmune Hemolytic Anemia
RBC destruction (Table 11-2).12-17
1. Corticosteroids The onset of the response is usually rapid.
a. Dosages and outcomes Significant hematologic improvement is often evident
b. Mechanisms of action within a few days, and most patients demonstrate
c. Adverse effects benefit within 10–14 days.2,3 It is unusual for improve-
2. Splenectomy ment to become evident after 3 weeks of therapy; a
a. Indications lack of response after this length of time should be
b. Clinical response considered a therapeutic failure. The first indication of
c. Durability of responses
d. Mechanisms of response a response might be a transient but often substantial
e. Value of radiolabeled red cell studies in selection of patients rise in the reticulocyte count. The degree of reticulo-
for splenectomy cytosis attained reflects, in part, the pretherapeutic
f. Adverse effects reticulocyte level. The elevation is less striking when
3. Immunosuppressive Drugs the initial reticulocyte counts are high. The cause of
a. Early reports the reticulocytosis is uncertain; some investigators
i. Azathioprine, cyclophosphamide, chlorambucil, vinca have suggested that it results from temporary marrow
alkaloids
ii. Indications and therapeutic regimens
stimulation, while others have indicated that prema-
iii. Adverse effects ture marrow release or a delay in maturation of the
b. Newer immunosuppressive agents reticuloycte appears more likely.6
i. Cyclosporine Subsequently, the hemoglobin and hematocrit
ii. Mycophenolate mefetil levels increase, at which time the reticulocyte count
iii. Rituximab
falls. For those patients with severe enough hemolysis
4. Danazol
to require RBC transfusions, an early indication of
5. Intravenous Immunoglobulin (IVIG) response is a decrease in the transfusion requirement.
a. Mechanisms of action Although evidence of improvement often occurs
b. Clinical results quickly, the response might not be maximal until after
6. Plasma Exchange 3 weeks of therapy.4
7. Hematopoietic Stem Cell Transplantation Improvement of symptoms frequently occurs
a. Results in animal models
before a hematologic response is apparent. The
b. Resolution of autoimmune disease following allogeneic stem patient might experience less fatigue, an improve-
cell transplantation ment in strength, decreased malaise, disappearance of
c. Autologous transplants for autoimmune disease fever, and a sense of well-being. The findings, how-
d. Future directions ever, might be nonspecific results of corticosteroid
Cold Agglutinin Syndrome therapy and are not always associated with hemato-
1. Avoidance of cold logic improvement.
2. Corticosteroid therapy Serum bilirubin values are of less significance in
3. Immunosuppressive drugs monitoring the effects of therapy, and they could
a. Cyclophosphamide and chlorambucil
b. Fludarabine
decrease slowly after the hematocrit and reticulocyte
c. Rituximab counts indicate improvement. Although an improve-
d. Alpha-interferon ment in the LDH values is to be expected, surprisingly
4. Plasma exchange little information is available on the use of this test to
5. Splenectomy monitor the effectiveness of therapy.
6. Intravenous immunoglobulin
7. Danazol
Paroxysmal Cold Hemoglobinuria HIGH-DOSE CORTICOSTEROID THERAPY
1. Supportive care
2. Avoidance of cold High-dose corticosteroid therapy has been suggested
3. Corticosteroids as a promising means of treating patients, especially
4. Splenectomy those who are not responsive to standard doses.
Secondary Autoimmune Hemolytic Anemias Ozsoylu18,19 has reported the use of megadose methyl-
Chronic lymphocytic leukemia prednisolone for numerous hematologic disorders
Systemic lupus erythematosus and is a proponent of such therapy. He recommends a
Hodgkin’s disease regimen of long-term daily megadose methylpred-
Non–Hodgkin’s lymphoma and other lymphoproliferative disorders
Mycoplasma pneumoniae infections and infectious mononucleosis
nisolone intravenously as follows: 30 mg/kg/day for
3 days, then 20 mg/kg/day for 4 days; subsequently,
10, 5, 2, and 1 mg/kg/day for 1 week each. He
reported “complete cure” of four patients with
Management of Autoimmune Hemolytic Anemias 403

TABLE 11-2. INITIAL CORTICOSTEROID RESPONSE RATE IN IDIOPATHIC WARM


ANTIBODY AIHA

Total Number of Percent Reference


Authors (Year) Patients Responders Responders Number

Pirofsky (1969) 33 25 76 12
Allgood, Chaplin (1967) 44 37 84 13
Eyster, Jenkins (1969) 13 10 77 14
Dameshek, Komninos (1956) 21 20 95 15
Dacie (1962) 23 17 74 16
Horster (1961) 167 139 83 17
TOTAL 301 248 82

Evans’s syndrome using this regimen and states that been obtained, and it might be beneficial to continue
there is little toxicity. the same dose for 1 to 2 weeks after stabilization of the
Meyer and colleagues20 treated seven patients hemoglobin and hematocrit. Thereafter, a progressive
with previously untreated WAIHA with pulsed high- but slow reduction of dosage is indicated. The follow-
dose corticosteroids. Most patients received six ing schedule is recommended: a weekly reduction of
courses of 40 mg dexamethasone orally or intra- the daily dose of prednisone by 10 mg or 15 mg until a
venously for four consecutive days on 28-day cycles. dose of 30 mg per day is reached. Thereafter, a reduc-
Before dexamethasone administration, hemolysis tion by 5 mg of the daily dose is made every week or
was decompensated in all seven patients. An two until a dose of 15 mg per day is reached.
improvement in hemolysis occurred in all cases, Subsequently, a reduction by 2.5 mg every 2 weeks
although relapses were frequent. The authors did not should be employed, with the aim of reducing the dose
attempt to compare the response rate of high-dose to zero. Although one might be tempted to discontinue
therapy with standard doses. The high-dose regimen steroids more rapidly, recurrences seem more likely
was said to be well tolerated. unless patients are treated for a minimum of 3 or
Meytes and coworkers21 reported a 42-year-old man 4 months with low doses of corticosteroids following
who had acute hemolysis associated with a high- subsidence of an acute episode of hemolysis.10,23
thermal-amplitude anti-Pr IgG cold agglutinin. The Rosse8 recommends an initial dose reduction by
patient’s hemolysis did not respond to 60 mg of 10 mg per day every week until 20 mg per day is being
prednisone, and his hemoglobin fell to 4.3 g/dL. administered. He suggests that this dose be maintained
Intravenous methylprednisolone was administered, for 1 month. Thereafter, the dose should be reduced to
and the patient’s clinical condition improved steadily. 10–15 mg per day or less, preferably given on alternate
The dosage of oral prednisone was reduced gradually, days (see later) in an effort to decrease the harmful
and 12 months later, he was found to be well. effects of corticosteroids. Subsequent reductions in
Although methylprednisolone given intravenously dose should be at 1-month intervals by steps of
is the usual drug for this purpose, dexamethasone 2.5–5.0 mg in the dose given on alternate days.
might be equally effective.22 Some authors warn of If a relapse occurs, one should return to the last
significant adverse reactions that could include hyper- previous dose. In some patients, remission may be
tension, cardiac arrhythmias, hypokalemia, psychosis, maintained by relatively small doses of prednisone
and infections.22 (5–10 mg per day). If a dose greater than 15 mg per day
of prednisone is required to keep the hematocrit level
above 30%, the response should be considered inade-
SUBSEQUENT MANAGEMENT quate, and other therapeutic maneuvers should be con-
Sudden cessation of therapy often results in prompt sidered. Some have recommended that 10 mg of
relapse, and one of the more common errors of man- prednisone should be the maximum dosage acceptable
agement is to taper corticosteroid therapy quickly to for chronic therapy,24 as the long-term side effects of
zero once a remission of hemolysis has occurred. On corticosteroids are often “disastrous.”12
the other hand, continued high-dose corticosteroid
therapy could lead to significant detrimental side LONG-TERM RESULTS
effects. Thus, it is important to develop a general stra-
tegy for reduction of dosage and criteria for institu- If there is to be a recurrence of hemolysis after
tion of other modes of therapy. corticosteroid-induced remission, it usually develops
Most authors recommend a regimen similar to the gradually when the recommended schedule is used
following guidelines. The initial dose of prednisone for tapering the dose. A majority of patients will
should be continued until a satisfactory response has relapse, however.
404 Immune Hemolytic Anemias

Approximately 20%–30% of patients achieve a and it is more likely that such “cures” represent spon-
lasting remission with prednisone therapy, 50% taneous remissions occurring during suppression of
continue to require some form of low-dose mainte- the disease manifestations by corticosteroids.
nance prednisone therapy for months, and another Glucocorticoids circulate in blood, either in the free
10%–20% either do not respond to prednisone or form or in association with cortisol-binding globulin.
require unacceptably high doses.4 The free form of the steroid can diffuse readily
Zupanska and associates11 found that only 28 of 80 through the plasma membrane and can bind with
patients (35%) had a long-lasting improvement after high affinity to cytoplasmic glucocorticoid receptors.
having 60–80 mg prednisone as initial treatment. The The formation of the ligand-receptor complex is fol-
remaining patients needed additional or alternative lowed by its “activation” (that is, translocation into
treatment because of intolerance or complications of the nucleus and binding to what are called acceptor
corticosteroid therapy or an inadequate response. sites). The bound complex modulates transcription of
Older data reported by Dacie16 indicated that only 3 specific genes that encode proteins responsible for the
of 23 patients (13%) were apparently “cured,” and action of glucocortocoids.5
Allgood and Chaplin13 indicated that 7 of 44 patients In addition to modulating transcription, glucocorti-
(16%) achieved a complete and permanent remission coids also have effects on later cellular events, includ-
lasting from 9 months to 11 years following a single ing RNA translation, protein synthesis, and the
course of corticosterodid therapy. Dameshek and secretion rates of specific proteins.
Komninos15 reported that remission was maintained From a clinical standpoint, the most important
after discontinuing corticosteroids in 8 of 40 patients effect of glucocorticoids on B cells relates to antibody
(18.6%). Some of the patients who relapse can be production. Low doses do not affect serum immuno-
maintained on acceptably low doses of prednisone globulin levels or antibody synthesis in vivo, but a
(10–15 mg per day or less), but about 50% of the brief course of daily high-dose prednisone decreases
patients will require other forms of therapy.13 serum immunoglobulin levels, with maximal sup-
pression observed 2 to 4 weeks after treatment.5 This
suppression is the result of an initial increase in
ALTERNATE-DAY THERAPY immunoglobulin catabolism, which is followed by
decreased synthesis.
There is a disassociation between the plasma and bio-
A number of these mechanisms are particularly rele-
logical half-life of glucocorticoids. Thus, the duration
vant for patients with AIHA and are described next.
of action of a glucocorticoid is not determined by its
Reduction of Antibody Synthesis. Reduction of
presence in the circulation, but likely is due to intra-
antibody synthesis certainly plays a role in the devel-
cellular effects secondary to binding of the steroid to
opment of remission for most patients treated for
cytoplasmic receptors.25 Understanding of this
WAIHA. Glucocorticoids are known to reduce anti-
concept has allowed clinicians to lengthen the inter-
body production. Rosse26 used the C1 fixation and
vals between administrations of the drug to further
transfer test to study the quantitative immunology of
decrease steroid side effects.25 For this regimen to
immune hemolytic anemia and demonstrated that,
work, a relatively short-acting steroid such as pred-
when patients achieved remission as a result of corti-
nisone must be chosen; alternate-day therapy is more
costeroid therapy, concentration of serum antibody
likely to be successful if the total dose required is
fell to low levels.
modest (e.g., averaging about 15 mg/day). One may
Numerous other investigators have documented
convert to alternate-day therapy by gradually increas-
similar findings by serologic studies. That is, if a
ing the dose on the first day of each 2-day cycle while
patient derives substantial benefit from corticosteroid
decreasing the dose on the second day, or by doubling
therapy, this usually is reflected in an improvement in
the daily dose and abruptly discontinuing treatment
the serologic findings.16 The direct antiglobulin test
on the alternate day. An alternate-day regimen has not
(DAT) becomes less strong and eventually could
been shown to decrease the propensity for osteoporo-
become negative, and free antibody, if originally
sis or posterior subcapsular cataract formation, but
present, most often is no longer discernable. For
the other common side effects are said to be dimin-
example, Evans27 reported that therapy resulted in
ished or eliminated.25
serum antibody disappearing in three of seven
patients. The DAT became weaker in six patients and
MECHANISMS OF ACTION negative in one. Dameshek and Komninos15 reported
improvement in the DAT titer in 19 of 24 patients,
Several mechanisms have been documented as being although in only 1 patient did it become negative
responsible for the remissions induced by cortico- permanently. In five patients, however, the strength
steroids. The relative role of each mechanism is of the reaction was apparently unaltered despite
difficult to quantify and probably varies from patient clinical and hematologic improvement. Serum anti-
to patient. None of the described mechanisms offers body became weaker in all 19 patients in whom anti-
an adequate explanation for the minority of patients body was originally found and became undetectable
who develop complete and permanent remissions, in 8 patients.
Management of Autoimmune Hemolytic Anemias 405

Such serologic improvement frequently is under- in clearance of RBCs sensitized with both IgM and
estimated if only the DAT is monitored, and particu- IgG antibody. The decrease in clearance was more
larly if only a single dilution of an antiglobulin marked in the case of IgG-coated RBCs and was dose
serum is used. Changes in serologic findings are dependent. The activity of the corticosteroids was
much more obvious if DAT titrations are performed first manifest after 4 to 5 days of treatment and
and the titer of the serum antibody is determined. became maximal after 8 days. The authors advanced
Chaplin28,29 illustrated a case in which apparent the hypothesis that one of the in vivo effects of
descrepancy between clinical improvement and lack corticosteroids was to decrease the number and/or
of change in the antiglobulin test was resolved by avidity of macrophage receptors for IgG and C3.
antiglobulin test titrations. The DAT at an optimal These experiments correlate with clinical experience
dilution of antiglobulin serum was 4+ at the time indicating much greater effectiveness of cortico-
corticosteroids were begun, and it remained so after steroids in WAlHA than in hemolytic anemia caused
40 days of therapy. During this time, the 51Cr T50 by IgM cold antibodies.
improved from 7 days on initial measurement to Kay and Douglas31 also studied monocyte recogni-
near-normal levels. Reactions at other dilutions of tion of immunoprotein-coated RBCs in vitro in 21
the antiglobulin serum, however, were clearly and patients with AIHA. They found that a majority of
progressively weaker when measured 14 and 40 days patients exhibited enhanced interaction between their
after the onset of therapy. Table 11-3 illustrates these monocytes and autologous immunoprotein-coated
findings. RBCs, compared with normal monocytes exposed to
The favorable effect on the DAT or a diminution or the same immunoprotein-coated erythrocytes. In an
disappearance of free antibody in the serum of a suc- animal model, 3 to 5 days of corticosteroid therapy
cessfully treated patient occurs relatively slowly, and resulted in a decreased ability to remove IgG-
it might not be discernable within the first week of sensitized RBCs and/or complement-sensitized cells
treatment. Thereafter, the response, if it occurs, is from their circulation. Both splenic and hepatic recep-
usually relatively rapid and might be well marked tor activity were inhibited. Macrophage activation
by the end of the second week.16 Thus, to the extent had the opposite effect. Five days after the intra-
that serologic reactions reflect concentrations of venous administration of bacille Calmette-Guérin
serum and cell-bound antibody accurately, it appears (BCG), guinea pigs had a markedly increased rate and
that the suppression of antibody synthesis is not magnitude of clearance of IgG- and IgG/C3b-
likely to be the explanation for the very rapid sensitized RBCs.
improvement noted among many patients. More
likely, it is at least a contributing factor in long-term ADVERSE EFFECTS OF
control of AIHA. CORTICOSTEROID THERAPY
Altered Antibody Avidity. A second mechanism for
therapeutic effect might be an alteration of antibody The beneficial effects of systemic glucocorticoids must
avidity for antigen on the RBC surface. Rosse26 be weighed against potential complications (Table 11-4),
reported a decrease in the number of antibodies bound including their probability of occurrence and their
to the RBC and a concomitant increase in concentration degree of harm.5 Numerous articles attest to the risks
of antibody in the serum within 48 hours of adminis- involved in chronic corticosteroid use.5,25,32-34 In the
tration of corticosteroids in humans. This could be the early years of aggressive glucocorticoid therapy, the
most immediate effect following the administration of consequences of extended high doses of glucocorticoid
corticosteroids to patients with AIHA. therapy were catastrophic, including death from infec-
Altered Clearance of Antibody-Coated Red Cells. tion and cardiovascular complications. Although such
A third effect of corticosteroids might be to alter the extreme complications are less frequent today, side
rate of clearance of antibody-coated RBCs by the reti- effects are still commonplace and usually cannot be
culoendothelial system. Frank and associates30 have avoided completely. Indeed, some risk of complications
shown that corticosteroid treatment caused a decrease is inherent in the attempt to obtain the therapeutic

TABLE 11-3. CHANGES IN DIRECT ANTIGLOBULIN TEST REACTIVITY DURING RESPONSE TO THERAPY

Reciprocal of Dilutions of Antiglobulin Reagent

51Cr T50, Days 2 4 8 16 32 64 128 256 512 1024

Before treatment 7 3+ 4+ 4+ 4+ 4+ 4+ 4+ 4+ 4+ 3+
After 14 days of treatment 12 3+ 3+ 4+ 4+ 4+ 4+ 4+ 3+ 1+ 2+
After 40 days of treatment 20 2+ 3+ 3+ 4+ 4+ 3+ 2+ 1+ 1+ 0

Agglutination reactions are graded as 1+ to 4+.


406 Immune Hemolytic Anemias

diuretics, anabolic steroids, tibolone, calcitonin, and


TABLE 11-4. MEDICAL COMPLICATIONS OF GLUCO- bisphosphonates.25,32-34 These measures often lack evi-
CORTICOID THERAPY dence-based support, however.33 There is no evidence
for benefit from alternate-day treatment in the context
of osteoporosis prevention.33
The American College of Rheumatology Ad Hoc
Committee on Glucocorticoid-Induced Osteoporosis36
concluded that supplementation with calcium and
vitamin D at a dosage of 800 IU/day, or an activated
form of vitamin D (e.g., alfacalcidiol at 1 μg/day or
calcitriol at 0.5 μg/day) should be offered to all
patients who are receiving long-term low-to-
medium-dose glucocorticoid therapy and who have
normal levels of gonadal hormones. Although
supplementation with calcium and vitamin D alone
generally do not prevent bone loss in such patients,
supplementation with calcium and an activated form
of vitamin D will prevent bone loss. Additional
recommendations were made for other classes of
patients.
Avascular Necrosis. Avascular necrosis is the
second major bone complication of corticosteroids.
The most common site is the femoral head, although
it can involve a number of other sites including
humeral heads, femoral condyles, tibial plateaus, and,

TABLE 11-5. ADVERSE EFFECTS OF


CORTICOSTEROIDS
From Boumpas DT, Chrousos GP, Wilder RL, Cupps TR, Balow JE:
Glucocorticoid therapy for immune-mediated diseases: Basic and clincal Organ System Condition
correlates. Ann Intern Med 1993;119:1198–1208.
Definite Adverse Effects of Low-Dose Corticosteroids
Eye Cataracts
benefits of glucocorticoids. The risks of adverse effects Glaucoma
Endocrine Diabetes mellitus
of corticosteroids are theoretically dose- and time- Skin Acne
dependent.5 Nevertheless, even with low-dose corti- Hirsutism
costeroids, significant adverse effects can occur Purpura
(Table 11-5).35 Skin atrophy
Osteoporosis. Corticosteroids appear to exert both Spontaneous skin tearing
direct and indirect actions on bone and calcium Probable Adverse Effects of Low-Dose Corticosteroids
homeostasis, leading to osteopenia. The pathogenesis Musculoskeletal Osteoporosis
of osteoporosis is complex, but the key factor is an Myopathy
imbalance between bone growth and resorption.5 A Withdrawal symptoms
Endocrine HPA dysfunction
general consensus exists that osteoporosis and its
associated fractures are the most serious side effects of High-Dose Corticosteroid Adverse Effects That Probably Do Not
long-term corticosteroid therapy.34 Although the rela- Occur with Low-Dose Corticosteroids
tive risk of osteoporosis among corticosteroid-treated Musculoskeletal Osteonecrosis
patients is unknown, it is estimated that 11% to 20% of Gastrointestinal Peptic ulcer
Pancreatitis
patients treated for more than 1 year with a dose of Bowel rupture
prednisone between 7.5 mg and 10 mg per day will Diverticular rupture
develop skeletal fractures.25 The most common sites of Cardiovascular Hypertension
fracture are the spine, ribs, and pelvis. Individuals at Lipoprotein abnormalities
Atherosclerosis
greater risk tend to be older than 50 years of age or Reproductive Fetal wastage
less than 15 years of age, are postmenopausal females, Fetal malformation
have a slim build, or have limited mobility. Neurologic Psychoses
In recent years, a wide variety of preventive mea- Cutaneous Delayed wound healing
sures and therapies have been proposed, including General Infections
hormone replacement therapy, calcium supplements Modified from Caldwell JR, Furst DE: The efficacy and safety of low-dose
administered with vitamin D metabolites, thiazides, corticosteroids for rheumatoid arthritis. Semin Arthritis Rheum 1991;21:1–11.
Management of Autoimmune Hemolytic Anemias 407

more rarely, the carpal, tarsal, metacarpal, and be associated with unpleasant dreams; thus, the steroid
metatarsal bones. Although traditional teaching has should be given as a single morning dose.32
suggested that avascular necrosis occurs predomi- Pediatric Complications. Glucocorticoids may
nantly among patients treated for prolonged periods retard bone age and arrest long bone growth, resulting
of time with high-dose corticosteroids, the condition in growth suppression. A dose of prednisone greater
is reported in a variety of clinical situations in doses than 5 mg per day may suppress growth in a child
used for short periods of time. In contrast to osteo- with a surface area of 1 m2. Although catch-up growth
porosis, avascular necrosis tends to occur in active may occur before puberty, this becomes impossible
patients. Treatment of early-stage avascular necrosis once the epiphyses close. Normal or near-normal
usually involves medical management and surgical growth patterns occur among patients on alternate-
core decompression. Advanced stages usually require day therapy.25
total joint replacement.25 Abnormalities of Glucose and Lipid Metabolism.
Susceptibility to Infection. The propensity of glu- A transient glucose intolerance that reverts to the pre-
cocorticoids to cause an infection diathesis is contro- treatment state despite continued therapy can
versial. In a meta-analysis of 71 trials involving more develop. Glucocorticoid-induced glucose intolerance
than 2000 glucocorticoid-treated patients, the rela- usually appears early in therapy and rarely is associ-
tive risk for infection across a number of clinical ated with serious complications.25
settings was approximately two times that of con- Additional Complications. Additional potential
trols.37 The risk varied according to the type of complications are listed in Tables 11-4 and 11-5.
disease being treated.
Gastrointestinal Complications. A prospective
study of 1400 patients receiving corticosteroid therapy
Splenectomy
showed an overall twofold increased risk for peptic INDICATIONS
ulcer disease. Multivariate analysis, however, showed
that concomitant use of nonsteroidal anti-inflammatory Splenectomy has a distinct advantage over other
drugs was the variable that explained the increased therapeutic options in that it offers the potential for
risk.38 Other risk factors that have been implicated are complete and long-term remission, and available data
a previous history of peptic ulceration and a total dose suggest that it does so for more than 50% of patients.2
of more than 1000 mg of prednisolone. For these This is in distinct contrast to corticosteroids, which
patients, prophylaxis with an H2-antagonist may be generally cause only a temporary suppression of
considered.25 AIHA, and to therapeutic maneuvers such as intra-
Luo and colleagues39 studied the potential risk venous immunoglobulin (IVIG), plasma exchange,
factors leading to peptic ulcer disease among 138 Danazol, immunosuppressive drugs, etc., which yield
autoimmune disease patients on corticosteroid treat- significant and long-lasting benefits for only a small
ment. They concluded that old age, smoking, and the percentage of patients. Accordingly, splenectomy
use of nonspecific cyclo-oxygenase inhibitors were should be considered for patients who do not respond
risk factors in these patients. adequately to corticosteroids according to the pre-
Hematologic Alterations. The total white blood cell viously stated guidelines.
count is increased in patients on corticosteroids. In practice, therapies with lesser benefit are often
Polymorphonuclear leucocytes are increased, whereas tried prior to splenectomy because some have minimal
lymphocytes (especially T cells), monocytes, and serious morbidity (e.g., IVIG, plasma exchange, and
eosinophils are decreased.32 Danazol), whereas there is a potential for serious mor-
Ophthalmic Complications. Systemic glucocorti- bidity and mortality in a small percentage of patients
coids may induce posterior subcapsular cataracts in as after splenectomy. Medical judgements vary regarding
many as 30% of patients. Most studies suggest that the place of splenectomy in individual cases. The fol-
cataracts rarely occur among patients treated with lowing is a review of the benefits and risks of the pro-
doses of less than 10 mg per day for 1 year. Such cedure, which is intended to provide an adequate
cataracts are usually bilateral and not associated with background for making such judgements.
altered visual acuity.25
Patients with increased intraocular pressure—includ- SURGICAL TECHNIQUE
ing patients with diabetes mellitus, myopia, and a
positive family history of glaucoma—may develop There has been a great deal of interest in the laparo-
increased intraocular pressure on oral glucocorticoids. scopic approach to splenectomy, which is emerging as
The elevated pressure is usually, but not always, the procedure of choice for splenic removal in patients
reversible if steroid therapy is discontinued.25 with hematologic disorders and normal to moderately
Central Nervous System Complications. Alterations enlarged spleens.40-49 The laparoscopic approach to
of behavior, including psychosis, agitation, insomnia, splenectomy hastens postsurgical recovery by reduc-
and depression, may be induced by systemic glucocor- ing pain and improving pulmonary function, leading
ticoids, and this effect might be directly correlated to to diminished hospital stay and reduced disability.
dose.25 Sleep disturbance is well recognized and could With accumulating data and increased experience,
408 Immune Hemolytic Anemias

laparoscopic splenectomy is emerging as the gold in Table 11-6.13,16,54-71 These data indicate that splenec-
standard for the management of various hematologic tomy appears to offer significant benefit in about 60%
disorders.40,41 of patients.
More recently, Coon72 reported the results of splenec-
CLINICAL RESPONSE tomy in 113 patients with hemolytic anemia, 52 of
whom had AIHA. Thirty-three of the patients with
The beneficial effect of splenectomy in acquired AIHA (63%) had an excellent response (steroid therapy
hemolytic anemia seems to have been reported for the terminated during the postoperative period and hema-
first time by Micheli in 1911.16 He described a 22-year- tocrit stable at 30% or greater). The mean follow-up
old man with hemolytic anemia with the probable was 33 months, although 14 of these patients had been
etiology of an autoimmune disease; a preoperative followed for less than 2 years. In addition, 11 patients
RBC count was 1.8 million/μL, which increased to (21%) were improved (hematocrit maintained at 30% or
4.5 million/μL one month after surgery. above while the patient is receiving 15 mg/day or less
In 1913, a good response to splenectomy was of prednisone or equivalent); the mean follow-up was
reported by Antonelli50 for a 40-year-old woman 73 months. Only eight patients (15%) failed to demon-
with hemolytic jaundice. The preoperative RBC strate improvement. The splenectomies were per-
counts had fluctuated between 715,000 and formed without mortality and with minimal morbidity.
2,000,000/μL; 5 months after surgery, the RBC count The authors cautioned that a portion of these patients
was 4.8 million/μL. Antonelli suggested that, might relapse at a later date.
although the spleen was of pathogenic importance, Of the patients reported by Coon, only four had an
hemolysis might be present in other places in the underlying disease (lymphoma or CLL); three
body and that splenectomy might act primarily by achieved an excellent and sustained response during
eliminating a source of destruction of RBCs. In 1920, periods of observation of 32–45 months. Ten patients
Eppinger51 described congenital and acquired forms had an associated thrombocytopenia (Evans’s syn-
of “hemolytic icterus” and was a strong advocate for drome), and in eight of these the platelet count
splenectomy. promptly returned to normal values after splenec-
Other favorable accounts followed, and by 1940 tomy; one patient who had a recent splenectomy
Dameshek and Schwartz52 were able to collect reports showed a rise in platelet count that had not yet
of 23 patients with acute hemolytic anemia (including reached normal values by the time of publication.
4 patients of their own), 20 of whom responded favor- Eight patients had DAT-negative hemolytic anemia.
ably to splenectomy. Later, Dameshek53 reported good Four had an immediate and excellent postoperative
results in 10 of 18 patients he studied personally. response. Three had a slow increase in hematocrit
Reports concerning the effectiveness of splenec- over a 6-month period and have remained in
tomy published between 1948 and 1974 are tabulated complete remission without steroids. The eighth

TABLE 11-6. RESULTS OF SPLENECTOMY IN IDIOPATHIC WARM ANTIBODY AUTOIMMUNE HEMOLYTIC ANEMIA

Authors Year Number of Cases Number Improved Percent Improved Reference

Stickney and Heck 1948 22 13 59 54


Robson 1949 9 7 78 55
Welch and Dameshek 1950 34 17 50 56
Dreyfus et al. 1951 5 2 40 57
Learmonth 1951 15 7 47 58
Chertkow and Dacie 1956 21 12 57 59
Young et al. 1957 18 12 67 60
Crosby and Rappaport 1957 27 17 63 61
Dausset and Colombani 1959 31 8 26 62
Dacie 1962 26 15 58 16
Goldberg 1966 13 11 85 63
Kinlough et al. 1966 9 8 89 64
Allgood and Chaplin 1967 25 18 72 13
Mappes and Fischer 1969 12 3 25 65
Schwartz et al. 1970 7 6 86 66
Devlin et al. 1970 5 4 80 67
Ahuja et al. 1972 4 4 100 68
Christensen 1973 13 12 92 69
Habibi et al.* 1974 16 12 75 70
Ikkala et al. 1974 4 3 75 71
Totals 316 191 60

* Children
Management of Autoimmune Hemolytic Anemias 409

patient had been operated on recently and was main- Hemoglobin response
taining a satisfactory hematocrit during the tapering
15
of steroid therapy.
Wilhelm and colleagues73 reported on the results of

Mean hemoglobin (g/dL)


400 splenectomies performed for hematologic disor-
ders between 1963 and 1982, only 8 of which were per- 10
formed for AIHA. For these eight patients, the mean
preoperative hematocrit was 26% (range, 19–35%), Idiopathic WAIHA
compared with a mean postoperative hematocrit at
5 Secondary WAIHA
2 to 4 weeks of 44% (range, 30–53%). No information
about long-term follow-up was provided. No mortal-
ity was associated with the surgery.
Ly and Albrechtsen74 reported on the results of
0
splenectomy for 221 adult patients with hematologic At splenectomy 2 mo 6–12 mo
disorders, 15 of whom had idiopathic WAIHA. FIGURE 11-1. Hemoglobin levels before and after splenectomy in
“Complete and lasting remission” was achieved for patients with idiopathic WAIHA (open square) and those with
nine patients (60%), partial remission for three (20%), systemic disease associated WAIHA (solid square). Data are means
no remission for two (13%), and one patient was not with one standard error. (From Akpek G, McAneny D, Weintraub L:
evaluable. The duration of follow-up was not indi- Comparative response to splenectomy in Coombs-positive autoimmune
hemolytic anemia with or without associated disease. Am J Hematol
cated. In the single patient with cold reactive antibo- 1999;61:98–102.)
dies, a partial remission was obtained by splenectomy.
The only postoperative death was caused by septi-
cemia after reoperation for massive bleeding in a patients with refractory idiopathic AIHA but that the
patient treated preoperatively with heparin. procedure should be considered “cautiously” for
Akpek, McAneny, and Weintraub75 compared the patients with secondary AIHA. Nevertheless, 41% of
results of splenectomy for 12 patients who had the patients with secondary AIHA who were refractory
idiopathic WAIHA with the results for 18 patients to medical therapy maintained an adequate hemoglo-
who had “secondary AIHA” associated with CLL, bin level after splenectomy.
non–Hodgkin’s lymphoma, systemic lupus erythe- There was one postoperative death among patients
matosus, mixed collagen vascular disease, and with idiopathic AIHA, involving a patient who
myelofibrosis (Table 11-7) (Figure 11-1). Nine of 11 underwent splenectomy during emergency surgery
(82%) evaluable patients with idiopathic AIHA and for a perforated ulcer while receiving steroid therapy.
3 of 16 (19%) patients with secondary disease achieved The authors stated that this fatal complication was
a complete response (normal hemoglobin with no most likely due to ulcer perforation rather than to
additional therapy lasting for at least 6 months). splenectomy per se. One patient with advanced
Among patients with idiopathic AIHA, the normaliza- non–Hodgkin’s lymphoma and one with CLL died of
tion of hemoglobin levels occurred at a median of sepsis in the postoperative period.
2 months after surgery. Partial responses (at least 50% Rosen and associates47 reported the outcome of
improvement in hemoglobin with or without additional laparoscopic splenectomy, which is now the proce-
treatment) were obtained in an additional 18% of dure of choice for elective splenectomy at the
idiopathic and 37% of secondary cases, respectively. Cleveland Clinic Foundation. They reported on the
Both the complete response and the overall response results of 147 surgeries for benign and malignant
rates were statistically different between the two groups hematologic disorders. Seven patients (5%) required
(P = 0.001 and 0.02, respectively). The authors con- conversion to open splenectomy, and postoperative
cluded that splenectomy is an effective treatment for complications occurred in 23 patients (26%). Eight of
the nine patients (89%) with AIHA had a remission of
their disease. The authors concluded that laparoscopic
splenectomy is safe and efficacious for a multitude of
TABLE 11-7. CLINICAL RESPONSE TO benign and malignant hematologic indications.
SPLENECTOMY

WAIHA with DURABILITY OF RESPONSES FOLLOWING


Idiopathic WAIHA Associated SPLENECTOMY
Response (n = 11) Disease (n = 16)
Remissions are not always permanent. For example,
Complete response 9 (82%) 3 (19%)
Partial response 2 (18%) 6 (37%)
Allgood and Chaplin13 reported that 10 patients had
No response None 7 (44%) a complete and permanent hematologic remission
Median follow-up (months) 18 (2–107.5) 10.9 (2–188) during a 1- to 10-year period of follow-up, but
6 other patients relapsed from 4 months to 8 years
From Akpek G, McAneny D, Weintraub L: Comparative response to
splenectomy in Coombs-positive autoimmune hemolytic anemia with or without after splenectomy. Zupanska and coworkers11
associated disease. Am J Hematol 1999;61:98–102. described a patient who, following splenectomy,
410 Immune Hemolytic Anemias

remained in full clinical reemission without any emphasized in reviews of the role of the spleen in
treatment for 15 years but then developed several hemolytic disorders, factors of significance include
relapses over the next 13 years. the characteristics of particulate flow through the
Detailed statistics regarding relapse rates after an microvasculature, the environmental characteristics
initial remission after splenectomy have not been specific to the spleen, the deformability and plasticity
reported systematically. Nevertheless, in spite of the of the red cell, and the highly specific cell-to-cell inter-
ever-present possibility of recurrence, Coon72 reassur- actions between RBCs and tissue cells.78,79
ingly reported that excellent responses or improvement
were maintained during median follow-up periods of
PREDICTION OF RESPONSES TO SPLENECTOMY
33 and 73 months, respectively.
If an incomplete remission develops or a relapse The Value of Radiolabeled Red Cell Studies in
occurs after splenectomv, much lower doses of corti- the Selection of Patients for Splenectomy. The
costeroids might prove effective in controlling the introduction of 51Cr as a RBC label was followed
disease activity.29 Christensen69 reported that the shortly by the suggestion that surface detection of the
prednisone requirement had decreased for 12 of 13 radioactivity related to specific organs could be used
patients who had undergone splenectomy. to determine the predominant sites of red cell destruc-
tion.80-82 Numerous subsequent studies have applied
MECHANISMS OF RESPONSE TO SPLENECTOMY this technique to hemolytic anemias.63,68,69,83-91
Technical difficulties have been emphasized regard-
As with corticosteroids, splenectomy probably pro- ing collination of the apparatus, marking of the
duces benefit by more than one mechanism. One mech- surface sites for counting, and analysis of the data.79,89
anism is decreased production of antibody. Although it In addition, the contribution of the RBC pool to the
may appear illogical that the removal of a small portion surface detectable radioactivity has produced a com-
of the reticuloendothelial system should result in the plication that has seldom been explicitly addressed in
disappearance of RBC autoantibodies, this is indeed the analysis of results.79 Clinical studies have led to
the case in a significant number of patients. Dacie16 various degrees of enthusiasm for the technique.
emphasizes that in patients who improve, the serologic Three major deterrents exist that prevent ready
signs of the disease will become less marked. If anti- acceptance of the thesis that splenic sequestration
body had been detected in the serum before splenec- studies with 5ICr-labeled RBCs should have a promi-
tomy, it will probably disappear, and the intensity of nent role in determining whether to perform splenec-
the DAT will lessen. Indeed, the DAT could become tomy on patients with AIHA. First, Nightingale and
completely negative in patients with complete hemato- coworkers92 point out that many predictions of the
logic remission, although it often remains positive for value of splenic sequestration studies in hemolytic
years as the only sign of autoimmunization. Dacie16 anemia have been largely based on (and extended
also reported that the DAT became negative in five of from) the findings in hereditary spherocytosis, in
seven patients who had clinical cures of WAIHA by which splenectomy is always successful. Second, the
splenectomy. Allgood and Chaplin13 did follow-up concept that measurement of splenic sequestration of
antiglobulin tests for eight patients who had a complete radiolabeled RBCs can predict the outcome of
remission, and the results were negative in four of splenectomy ignores the fact that the decreased pro-
them. Habibi and coworkers70 described eight patients duction of antibody as a result of splenectomy is a
who were cured of hemolysis after splenectomy, and major mechanism by which the operation produces
the serologic test results became negative in five of clinical benefit.13
these patients. Third, Allgood and Chaplin13 emphasize that few
The diminution in antibody production after patients have been reported who have undergone
splenectomy in patients with AIHA is consistent with splenectomy when external organ scanning failed to
the report of Karpatkin and associates,76 who studied indicate splenic sequestration. For example, Ahuja68
the effect of splenectomy in a similar syndrome, idio- selected patients for surgery mainly on the basis of
pathic autoimmune thrombocytopenic purpura. They surface counting. No patients who showed evidence
demonstrated a significant decline in serum anti- of RBC destruction in the liver alone or in whom there
platelet antibody titer in seven out of eight patients. was no excess accumulation in either liver or spleen
McMillan and colleagues77 also reported that the underwent the operation. Similarly, Kinlough and
spleen is an important site of antiplatelet antibody colleagues64 concluded that studies with 51Cr-labeled
production. RBCs could lend valuable assistance in the selection
A splenectomy also results in the removal of a of patients for operations, but all patients in their
potent site of RBC damage. Thus, the same degree of series had a “splenic uptake” sequestration pattern.
red cell sensitization may result in less shortening of Goldberg63 selected 13 patients for splenectomy on
red cell survival time. Rosse26 estimated that after the the basis of an elevated spleen-to-liver ratio and con-
spleen is removed, up to 10 times as much antibody as cluded that results based on this criterion are better
was present before splenectomy must be present on than among randomly selected patients reported in
the red cell to cause the same degree of lysis. As the literature.
Management of Autoimmune Hemolytic Anemias 411

Indeed, a significant number of patients with prediction of failure by the isotope study. In cases in
WAIHA have had favorable responses to splenec- which the indications for splenectomy are not com-
tomy in spite of 51Cr sequestration studies that pre- pelling (e.g., an incomplete remission maintained on
dicted a therapeutic failure. For example, Allgood less than 15 mg of prednisone per day, or patients
and Chaplin13 reported a good response to splenec- with relative contraindications to splenectomy), the
tomy in four of seven patients who failed to show sequestration study might be of some value in arriv-
evidence for splenic sequestration. Parker and associ- ing at a decision. Among those who require 10–15 mg
ates87 studied 12 patients and found that three of per day or more of prednisone for adequate control of
seven patients with spleen-to-liver ratios of less than hemolysis, however, no patient should be denied the
2.5 had a good result from splenectomy, while two of possible beneficial results of splenectomy on the basis
five patients with ratios greater than 2.5 had a poor of a 51Cr splenic sequestration study.
response. They concluded that surface counting Serologic Tests as a Predictor of Efficacy of
measurements are not reliable indicators of the Splenectomy. A number of investigators have
outcome of splenectomy. Ben-Bassat and colleagues83 attempted to correlate the results of splenectomy with
reported that in three out of five patients, the the presurgical serologic findings in cases of warm
response to splenectomy was contrary to that antibody AIHA. Zupanska and colleagues11 stated
expected from sequestration studies. that excellent results were more frequent in patients
Zupanska and coworkers11 reported the results of who had no complement on their RBCs. The authors
splenectomy in comparison with the spleno-hepatic added, however, that the number of cases is too small
index and also in comparison with spleen weight to form firm conclusions.
(Fig. 11-2).11 These authors concluded that there was Similarly, neither the strength of the DAT nor the
no correlation between postsplenectomy results and presence or absence of antibody in the serum have
spleen-to-liver radioactivity ratios. Discrepant results been of predictive value. Although the quantity of
also have been reported by other investigators, and surface antibody has been shown under experimental
Crosby93 succinctly concludes that the test gives false- conditions to be an important determinant of red cell
positive and false-negative results, which vitiate its destruction, this is not measured reliably in quantita-
value.63,68,90 tive terms by the antiglobulin test.26,96 In 1976,
Therefore, clinical findings rather than 51Cr seques- Bowdler79 suggested that if techniques of investiga-
tration studies should indicate the advisability of per- tion would become more available regarding both the
forming splenectomy.12,13,78,93-95 The operation need qualitative and quantitative characteristics of
not be considered if satisfactory remission results immunoglobulin bound to the RBC surface, it would
from corticosteroid administration. If corticosteroids reasonably be anticipated that decisions about
yield unsatisfactory results, the options are few: splenectomy would become more precise. This poten-
splenectomy, immunosuppressive drugs, and a tial has not been realized in the more than a quarter-
variety of miscellaneous therapies that will be century since his remarks were published.
reviewed shortly. The favorable results of splenec-
tomy are more impressive than those of any of the ADVERSE EFFECTS OF SPLENECTOMY
alternatives, and although a splenic sequestration
study might predict a higher probability of response Surgical Morbidity and Mortality. The mortality
than would be true for a randomly selected patient, a and morbidity associated with splenectomy vary
good clinical response frequently occurs in spite of greatly, depending on the underlying disease for which

Ratio g × 1000

4 4

3 3
Splenohepatic index

FIGURE 11-2. Results of splenectomy in comparison with


spleno-hepatic index and spleen weight. (From Zupanska B,
Sylwestrowicz T, Pawelski S: The results of prolonged treatment
Spleen weight

of autoimmune haemolytic anaemia. Haematologia 2 2


1981;14:425–433.)

1 1

Excellent Good Fair Poor Death


412 Immune Hemolytic Anemias

the operation is performed. Thus, Schwartz and associ- drome a true medical emergency. Severe hypoglycemia
ates66,97 reported on a series of 200 splenectomies for might also be present. The subsequent clinical course
hematologic disorders in which a 7% mortality often mirrors that of the Waterhouse-Friderichsen syn-
occurred during the immediate hospitalization. All drome, with bilateral adrenal hemorrhages noted at
deaths, however, occurred in patients with idiopathic autopsy. Peripheral gangrene requiring amputations
thrombocytopenic purpura, thrombotic thrombocy- has been reported in survivors, secondary to the
topenic purpura, lymphomas, or leukemias. The imme- combination of hypotension and disseminated intra-
diate cause of death was usually cerebral hemorrhage vascular coagulation.
caused by severe thrombocytopenia. In good risk Despite appropriate antibiotics and intensive thera-
patients who underwent elective splenectomy, no peutic intervention, the overall mortality in older pub-
deaths occurred. Similar experience with almost identi- lished studies for established cases of OPSI varied
cal mortality rates has been reported in other large from 50% to 70%.108,110-112 More recent information
series.98-100 suggests that when informed patients seek medical
Postoperative morbidity occurred in 18% of the attention promptly, mortality could be reduced to the
patients reported on by Schwartz and colleagues.66 range of 10%.109 Of those patients who die, more than
The most frequent complication is that of left lower 50% die within the first 48 hours of hospital admis-
lobe atelectasis. Wound complications, septicemia, sion. Among patients who survive, other sequelae
and development of subphrenic hematoma and have included deafness associated with either menin-
abscess occurred almost exclusively in patients with gitis or mastoid osteomyelitis, and aortic insufficiency
myeloid metaplasia or hematologic malignancies with secondary to endocarditis.110
significant bleeding from the splenic bed. Responsible Organisms. Most instances of serious
Overwhelming Postsplenectomy Infection (OPSI). infection are due to encapsulated bacteria, such as
Morris and Bullock in 1919101,102 were first to predict Pneumococcus (Streptococcus pneumoniae). Other, less
that removal of the spleen would result in an increased common organisms include Haemophilus influenzae,
susceptibility to infection. King and Schumacher103 Neisseria meningitides, and Escherichia coli. Additional
reported in 1952 on five cases of fulminant sepsis in organisms that may cause OPSI include, but are not
splenectomized infants with hereditary spherocytosis. limited to, Plasmodia (malaria), Babesia, and Capnocyto-
In 1957, Smith and colleagues104 presented his data on phaga canimorsus (an organism acquired from animal
postsplenectomy infection in patients with thalassemia, bites).
and in 1960, Lucas and Krivit105 published data on Incidence and Risk Factors. The precise incidence
overwhelming infection in children following splenec- of OPSI remains controversial, and published esti-
tomy. It was not until the 1970s, however, that a distinct mates vary considerably.109 Lynch and Kapila108
syndrome became well defined and accepted, largely reviewed all published series and, in 1996, indicated
as a result of reviews by Singer106 in 1973 and that the lifetime risk for OPSI was 5%. Children less
Krivit102,107 in 1977 and 1979. Subsequently, a large than 15 years of age have a greater overall risk of OPSI
body of data has accumulated concerning the inci- (0.13%–8.1%) compared with adults (0.28%–1.9%). In
dence, prognosis, prevention, and management of this one retrospective review of 5902 postsplenectomy
entity, and the syndrome of overwhelming post- patients studied between 1952 and 1987, the incidence
splenectomy infection has been anointed with its own of OPSI was 4.4% in children under 16 years and 0.9%
acronym, OPSI. Despite the large number of published in adults.113 Another study calculated the risk of
cases, however, insufficient data exist to determine the sepsis as 0.73 per 1000 person-years after splenectomy
true incidence, exact nature of infection, risk factors, for hereditary spherocytosis.114 Schwartz and cowork-
and the contribution of underlying conditions.108 ers115 estimated the risk of fulminant infection as rea-
Clinical Findings. The initial prodrome may be sonably low—in the range of one case per 500
mild and nonspecific, with flulike symptoms that person-years of observation. The overall cumulative
include fever, malaise, myalgias, headache, vomiting, risk of infection severe enough to require hospitaliza-
diarrhea, and abdominal pain.109 The presence of gas- tion, however, was 33% by the end of a 10-year follow-
trointestinal symptoms should never prevent the up. Cullingford and associates116 reported that the
attending physician from entertaining a possible diag- incidence of overwhelming infection after splenec-
nosis of OPSI. Although meningitis or pneumonia tomy was only 0.04 per 100 person years.
accompanies OPSI in approximately 50% of cases, in Most of the published data antedate the widespread
many patients there is no obvious site of bacterial col- availability of the pneumococcal and Haemophilis
onization. When sufficient detail has been reported in influenzae vaccines, so current rates could be lower.
the literature, many patients will have had true rigors For instance, a Danish study found that the incidence
for a day or two before receiving definitive medical of pneumococcal infection in splenectomized children
management. decreased dramatically following the introduction of
This prodrome may be followed by rapid evolution the pneumococcal vaccine and the promotion of early
to full-blown bacteremic septic shock accompanied by penicillin therapy for febrile episodes.117
hypotension, anuria, and clinical evidence of dissemi- In addition to age, major factors relating to risk
nated intravascular coagulation, thus making this syn- include the reason for splenectomy and the subsequent
Management of Autoimmune Hemolytic Anemias 413

time interval from splenectomy. Patients with tha- should be performed, especially in infants and chil-
lassemia or sickle cell disorders and patients with lym- dren. Standard laboratory tests and appropriate x-ray
phomas are at greatest risk.108 Although most infections studies are indicated, and the possibility of dissemi-
occur within the first 2 years after splenectomy, 33% of nated intravascular coagulation and septic shock
postsplenectomy pneumococcal infections and 42% of should be anticipated.109
OPSI occurred more than 5 years after splenectomy.108 Managing Patients with OPSI. Although there is
The data of Waghorn and Mayon-White118 indicate that currently no proof that early treatment prevents incipi-
OPSI occurs at any age and that it does not necessarily ent bacteremia from progressing to full-blown OPSI, the
occur within the first few years after splenectomy literature does support improved survival from an
(Table 11-8). OPSI has been reported as late as 59 years aggressive approach.111 Possible choices of empiric
after splenectomy.118 antimicrobial agents useful fo the initial treatment of
Clinical and Laboratory Diagnostic Testing. Initial OPSI are included in Table 11-9.109 Intravenous
diagnosis must rely on a high index of clinical suspi- penicillin therapy generally has been superseded by
cion for any febrile presentation in a splenectomized broad-spectrum antibiotic therapy active against
patient, because aggressive early management is penicillin-resistant pneumococci, β-lactamase–producing
thought to be critical. Diagnostic work-up should H. influenzae, and other possible organisms that are
never delay the initiation of empiric antibiotic not normally penicillin sensitive. Expert opinion from
therapy. an infectious disease specialist should be sought
A quick and often helpful initial test is the examina- urgently. Aggressive supportive care is necessary,
tion of the peripheral blood smear and buffy coat for including appropriate evaluation and any necessary
the presence of bacteria. Visualization of organisms on management of disseminated intravascular coagula-
the peripheral smear suggests a quantitative bacteremia tion and septic shock. The use of high-dose cortico-
of greater than 106 micro-organisms/mL (104 or more steroids remains controversial but has not been
greater than the usual bacteremia), which contributes demonstrated to be of benefit.108
greatly to morbidity and mortality.108 Preventive Strategies. Education, immunoprophy-
Blood cultures are usually positive within 24 hours laxis, and chemoprophylaxis are all considered
and should be performed to identify the pathogen and important in the prevention of OPSI (Table 11-10).119
guide subsequent therapy. Cultures should also be Patient education is a mandatory strategy in
obtained from other sites, and lumbar puncture attempting to prevent OPSI. Unfortunately, studies
have shown that 11%–50% of postsplenectomy
patients remain unaware of their increased risk for
serious infection or of the appropriate health pre-
TABLE 11-8. SPLENECTOMY IN OPSI CASES cautions that should be undertaken.109 Published
guidelines regarding immunizations suggest that
Ages of OPSI Cases pneumococcal vaccine should be given a minimum of
Age Group No. of Cases (Total = 42) 2 weeks before elective splenectomy to ensure an
optimal antibody response. If this is not practicable,
<2 years 1 the patient should be immunized as soon as possible
2–19 years 0 after recovery from the operation. Reimmunization of
20–29 years 5 asplenic patients is recommended every 5 to 10 years.
30–39 years 10
40–49 years 10 Although there is a paucity of data showing clear clin-
50–59 years 7 ical efficacy in asplenic patients, it is encouraging to
60–69 years 4 note that in one reported series of asplenic hematol-
70–79 years 4 ogy patients, only four episodes of pneumococcal
80–89 years 1
septicemia/meningitis were observed in more 200
Time Interval from Splenectomy to OPSI vaccinated individuals within a 13-year period, and in
Time Interval No. of Cases (Total = 40)* all four episodes, the infecting capsular type was not
included in the vaccine.120
<3 months 2 H. Influenza type B vaccine is also generally recom-
1–4 years 2 mended for all patients who have not already
5–9 years 3
10–19 years 15
received it.121 The overall efficacy and utility of this
20–29 years 8 vaccine is less clear than for pneumococcal vaccine,
30–39 years 6 however, as most adults already have antibody and
40–49 years 1 many infections involve non–type B strains.109 The
>50 years 2 need for reimmunization is unclear.
Unknown 1
The efficacy and importance of meningoccoccal
* Child with congenital asplenia and adult with severe splenic atrophy not vaccination in splenectomized individuals is un-
included. known, although it is often recommended.108,109 The
Modified from Waghorn DJ, Mayon-White RT: A study of 42 episodes of
overwhelming postsplenectomy infection: Is current guidance for asplenic current meningococcal vaccine covers types A, C,
individuals being followed? J Infect 1997;35:289–294. W135, and Y but misses other pathogenic strains.109
414 Immune Hemolytic Anemias

TABLE 11-9. EMPIRIC TREATMENT FOR SUSPECTED OVERWHELMING


POSTSPLENECTOMY INFECTION

Drug Adult Dosea Pediatric Dosea

Cefotaxime 2 g IV every 8 hr 25–50 mg/kg IV every 6 hr


Ceftriaxone 2 g IV every 12–24 hr 50 mg/kg IV every 12 hr
+/– Gentamycinb 5–7 mg/kg IV every 24 hr 2.5 mg/kg IV every 8 hr
or
+/– Ciprofloxacinb,c 400 mg IV every 12 hr
+/– Vancomycind 1–1.5 g IV every 12 hr 30 mg/kg IV every 12 hr
a Doses are for normal renal function and should be adjusted if creatinine clearance is reduced.
b Gentamycin or ciprofloxacin may be added if an enteric or urologic source of infection is suspected.
c Ciprofloxacin is not indicated for children.
d Vancomycin should be added when pneumococcus with high-level penicillin resistance is likely.
From Brigden ML, Pattullo AL: Prevention and management of overwhelming postsplenectomy infection—An update. Crit Care Med
1999;27:836–842.

Immunity appears to diminish over the course of a In contrast, Flegg121 suggests that beyond 5 years
few years, suggesting a requirement for frequent postoperatively in adulthood, by which time the risks of
boosters. overwhelming infection are much reduced, prophylaxis
Influenza vaccine is recommended yearly and should be reserved for only the most vulnerable sub-
might be of particular value to asplenic patients by groups, that is, those with malignant or hematological
reducing the risk of secondary bacterial infection.119 disease.
The American Advisory Committee on Immun- Perhaps the biggest problem in managing post-
ization Practices also recommends Neisseria meningi- splenectomy patients is the lack of implementation of
tides vaccine.122 the foregoing preventive strategies. Waghorn and
Most authorities recommend antibiotic prophylaxis Mayon-White118 reported 42 cases of OPSI. They
for asplenic patients. Some recommend that lifelong found that pneumococcal infection had caused at least
prophylactic antibiotics should be offered in all 37 of the 42 episodes, but only 12 patients had
cases—especially in the first 2 years after splenec- received pneumococcal vaccine. They concluded that
tomy—for all children up to age 16 and when there is much more needs to be done to ensure that asplenic
underlying impaired immune function.119 Tradition- patients are warned of the risks of infection and pro-
ally, a single daily dose of penicillin or amoxicillin has vided with appropriate prophylactic measures.
constituted the regimen of choice. With the develop- Similarly, Deodhar and colleagues123 reported that
ment of resistant organisms, however, antibiotics with among 184 patients who had had a splenectomy
broader activity have increasingly been utilized.109 during a 12-year period, 58% had not received advice
or prophylaxis against infection and only 36% had
received pneumococcal vaccination.
TABLE 11-10. GUIDELINES FOR PREVENTION OF OPSI In addition, for patients not allergic to penicillin, a
supply of amoxycillin should be kept available, and
• All splenectomized patients and those with functional the patient should be instructed in immediate self-
hyposplenism should receive pneumococcal immunization (a, b). medication should symptoms of fever, malaise, or
• Documentation, communication, and reimmunization require shivering develop. Others recommend cefotaxime or
attention (a, b).
• Patients not previously immunized should receive Haemophilus
ceftriaxone as empiric treatment for symptomatic
influenzae type b vaccine (a, b). patients who have been taking antibiotic prophy-
• Meningococcal immunization is not routinely recommended (b). laxis.109 In any case, the patient should also seek
• Influenza immunization may be beneficial (b). immediate medical help.
• Lifelong prophylactic antibiotics are recommended (oral Thrombocytosis. An additional potential compli-
phenoxymethylpenicillin or an alternative) (a, b).
• Asplenic patients are at risk of severe malaria (a). cation of splenectomy is the development of post-
• Animal and tick bites could be dangerous (a). splenectomy thrombocytosis and thromboembolism.
• Patients should be given a leaflet and a card to alert health Splenectomy is usually followed by a mild, symptom-
professionals to their risk of overwhelming infection (a, b). less thrombocytosis that reaches a peak at about the
• Patients developing infection despite measures must be given a
systemic antibiotic and urgently admitted to hospital (a, b).
end of the second week and gradually subsides within
3 months.124 Occasionally, however, postsplenectomy
a = Based on published evidence. thrombocytosis persists. Hirsh and Dacie124 studied
b = Expert opinion.
nb: There are no randomized controlled trials or case-controlled studies on this
the postsplenectomy platelet count in patients suffer-
issue. ing from various types of anemia and in hematologi-
From Working Party of the British Committee for Standards in Haematology cally normal persons. Persistent postsplenectomy
Clinical Haematology Task Force: Guidelines for the prevention and treatment
of infection in patients with an absent or dysfunctional spleen. Br Med J 1996; thrombocytosis was noted in all patients with
312:430–434. Reprinted with permission of the BMJ Publishing Group. continuing anemia after splenectomy, and the height
Management of Autoimmune Hemolytic Anemias 415

of the persistent thrombocytosis was closely related to dence of thromboembolic complications of 3.9% with
the severity of the anemia (P < 0.001). No such rela- increased platelet counts, compared with 1.3% in
tionship existed between the platelet count and hemo- patients who did not have thrombocytosis following
globin level in a comparable group of patients who splenectomy (the difference was not statistically
had not been subjected to splenectomy. Visudhiphan significant). On the other hand, Traetow127 reported a
and coworkers125 also found a significant negative 6% incidence of thromboembolism in 223 patients
correlation between hemoglobin level and platelet with postsplenectomy thrombocytosis, compared
count following splenectomy (Fig. 11-3).125 Hirsh and with 0.4% in 250 patients with postoperative platelet
Dacie124 suggested that in the presence of active counts of less than 400,000/μL. Both of these groups
hemopoiesis, anemia stimulates both thrombopoiesis noted that if the platelet count was greater than
and erythropoiesis, but that increased thrombopoiesis 1,000,000/μL, there was no further increase in throm-
does not result in persistent thrombocytosis unless the boembolic risk.
spleen is removed. They indicated that the persistence Coon and colleagues128 studied 86 patients who
of thrombocytosis after splenectomy can usually be underwent elective splenectomy and detected deep
predicted; this happens when anemia continues after vein thrombosis in five patients by using labeled
splenectomy in association with active hemopoiesis. fibrinogen and dye phlebography. In none of these
Hirsh and Dacie124 also pointed out that post- five patients, however did an elevation in platelet
splenectomy thromboembolism in association with count to 600,000/μL develop before or at the time of
postsplenectomy thrombocytosis has been reported development of the thrombosis. None of 21 other
after splenectomy, and they reported on five patients patients who had a rise in platelet count to greater than
in whom this complication occurred out of 80 patients 1,000,000/μL had evidence of venous thrombosis.
who had undergone splenectomy for hematologic Clearly, other factors are important in the etiology
disease. Four of the five patients had congenital of postsplenectomy thromboembolic complications.129
hemolytic anemias, but none of the patients in their A number of authors have indicated that the throm-
series had an acquired immune hemolytic anemia. boembolic risk is highest among patients with hyper-
The authors were careful to point out that many other splenism and myeloprolifertive disorders such as
patients in their series had comparable increases in chronic myelogenous leukemia, agnogenic myeloid
the postsplenectomy platelet count, which had been metaplasia, and polycythemia vera.129,130 Some
maintained for many years without development of authors found no substsantial increase in thrombo-
thromboembolism. embolic risk in patients without myeloproliferative
Indeed, a relationship between postsplenectomy disease.126,131
thrombocytosis and the incidence of thromboem- Bensinger and coworkers132 reported on four
bolism is not always found. Boxer126 reported an inci- patients (none of whom had AIHA) who developed
hemorrhagic complications associated with “spectac-
ular” thrombocytosis postsplenectomy. Therapy with
melphalan (L-phenylalanine mustard) effectively con-
trolled the abnormal thrombocytosis and the clinical
15 evidence of disease.
Coon and colleagues128 concluded that there was no
need for the routine administration of prophylactic
antithrombotic therapy in patients in whom post-
r2 = 0.64 splenectomy thrombocytosis develops.
10 Thus, patients who have undergone splenectomy
for AIHA may develop thrombocytosis if anemia per-
Hb gm %

sists, but there are no clear indications for treatment


unless hemorrhagic or thromboembolic phenomena
occur.
5
ABNORMAL LABORATORY TESTS
FOLLOWING SPLENECTOMY
Thrombocytosis
Pseudohyperkalemia may be encountered following
splenectomy as a result of the in vitro release of intra-
0 500 1000 1500 cellular potassium from platelets during clotting.133-135
Platelets × 103 per mm3
This can be avoided by using heparinized tubes with
plasma separated without delay.
FIGURE 11-3. The correlation between hemoglobin level (g/dL) and The reduction or absence of normal splenic function
platelet count in 29 splenectomized subjects. (From Visudhiphan S,
Ketsa-Ard K, Piankijagum A, Tumliang S: Blood coagulation and platelet can be recognized by certain hematologic changes.136,137
profiles in persistent post-splenectomy thrombocytosis: The relation- Some of these are nonspecific, such as a slight to mod-
ship to thromboembolism. Biomed Pharmacother 1985;39:264–271.) erate increase in the white cell count and platelet count.
416 Immune Hemolytic Anemias

Findings of Howell-Jolly bodies, pitted erythrocytes, wealth of information regarding the various drugs,
and target cells in the blood smear, however, are of their mechanisms of action, pharmacokinetics, effec-
greater diagnostic significance. For reasons that still tiveness, and adverse effects.142,144-146 Unfortunately,
remain unknown, the red cell surface area is however, the literature regarding the appropriate use
increased after splenectomy, causing buckling and of these drugs in the treatment of AIHA is meager and
target cell formation.138 Nuclear fragments that nor- consists largely of anecdotal case reports.
mally are removed in the spleen are present in circu-
lating red cells and are termed Howell-Jolly bodies.138 EARLY REPORTS
They are almost always present in the asplenic state,
but only 1 of 100 to 1 of 1000 RBCs is affected. A sen- Dacie6 has reviewed the early medical literature
sitive indication of hyposplenism is the appearance regarding the development and use of immunosup-
of pits or pocks on the cell surface.138a,b,139 They pressive alkylating and antimetabolite drugs for the
consist of submembraneous vacuoles and can be therapy for warm antibody AIHA. Some of the more
seen only in wet preparations of RBCs using direct informative of these early reports include that of
interference-contrast microscopy.139 Schwartz and Dameshek,147 who reported the results
Oxidative drugs may produce Heinz bodies even in of the use of antimetabolites in 14 patients. This series
normal individuals, but those RBC inclusions are included six idiopathic cases, two associated with
removed effectively by the spleen. After splenectomy, systemic lupus erythematosus (SLE), and single cases
they may be observed in supravital-stained blood associated with myxedema, hepatitis, chronic lym-
films. Nucleated RBCs, on the other hand, are seen phocytic leukemia, Dilantin ingestion, virus infection,
only rarely on blood films after splenectomy, except and rheumatoid arthritis. Two patients were treated
in patients with hemolytic disorders, in whom their exclusively with immunosuppressive drugs. Nine of
number may increase dramatically.136 the 14 patients experienced a beneficial effect of
Mature circulating RBCs containing persistent gran- therapy with either 6-mercaptopurine or thioguanine
ules of nonheme iron are called siderocytes, while at a dosage of 2.5 mg/kg. Of nine patients who were
nucleated red cells bearing iron-containing inclusions prior steroid failures, four had good responses, and
are designated sideroblasts. In both cases, the iron- one had a partial response to cytotoxic therapy.
containing particles are known as siderotic granules. Taylor148 reported on a patient with AIHA and
Siderocytes are best demonstrated with the Prussian possible Hodgkin’s disease who failed to respond to
blue staining technique and are not detectable on corticosteroids and splenectomy. Over the next
ordinary Wright-Giemsa–stained blood smears. A few 6 years, remissions of the hemolytic state were
siderocytes can be found in the blood of an asplenic induced with triethylene melamine, chlorambucil,
but otherwise normal person, whereas substantial and cyclophosphamide.
numbers may be found in asplenic individuals with Hitzig and Massimo149 treated three children with
an underlying hematologic disease such as sideroblas- AIHA with azathioprine at a dosage of 2.5 mg/kg in
tic or Heinz-body anemia.137 combination with steroid therapy. In one patient there
was a complete response, and two experienced a
partial response that permitted the reduction of
Splenic Irradiation steroid dosage.
Splenic irradiation can be an effective modality in Habibi and colleagues70 treated seven children with
controlling AIHA and is utilized most often when AIHA with azathioprine, four of whom obtained good
patients are too ill for splenectomy.140 Markus and responses.
Forfar141 described a 73-year-old male with AIHA Worlledge150 reported that 6 of 14 patients treated
unresponsive to corticosteroids, whose condition pre- with 75–200 mg of azathioprine daily had a good
cluded splenectomy. A course of splenic irradiation response, and 1 patient had a partial response. The
was given, and his hemoglobin concentration subse- duration of treatment ranged from 4 months to
quently rose to normal. 6 years.
Skinner and Schwartz151,152 reviewed 42 reported
cases of warm antibody AlHA. Most of the patients
Immunosuppressive Drugs were treated with a combination of azathioprine and
Since the publication of the first edition of this book,1 corticosteroids, and in many cases splenectomy was
there have been remarkable advances in the develop- performed before the azathioprine had been insti-
ment of immunosuppressive drugs. Indeed, the disci- tuted. All patients seemed to have failed to respond to
pline of human organ transplantation has made major corticosteroid treatment. Twenty of the 42 patients
strides because of significantly more effective (48%) were said to have improved with immunosup-
immunosuppressive drug regimens, and they have pressive therapy. Unfortunately, no objective criteria
been used increasingly for management of autoim- for improvement were listed in many cases, and the
mune diseases.142,143 response was often merely described as “beneficial.”
The new millennium seems to have inspired a A similar analysis of the literature by Mueller-
number of comprehensive reviews that provide a Eckhardt and Kretschmer153 reported a 45% incidence
Management of Autoimmune Hemolytic Anemias 417

of “good” response in 66 patients with AIHA treated VINCA ALKALOIDS


with cytotoxic drugs.
Johnson and Abildgaard154 reviewed the literature The therapeutic effects of vinca alkaloids in AIHA
relating to pediatric patients and described two addi- have not been investigated systematically.158 There
tional cases. In all, 9 of 17 patients (53%) showed have been some anecdotal reports of benefit after the
improvement. Eight of the nine children who use of vinblastine and vincristine, however.159,160
improved received azathioprine as one of their Vincristine was generally given as part of a multiple-
immunosuppressive agents, but at least four children drug regimen, often to patients with underlying lym-
who failed to improve also had received this drug. phoproliferative disorders.161-163
Dovat and associates164 reported an 8-month-old
male with X-linked lymphoproliferative disease who
SUBSEQUENT REPORTS underwent an unrelated, partially matched (with major
Moyo and associates155 studied high-dose cyclophos- mismatch at DR locus) cord blood stem cell transplant.
phamide (50 mg/kg ideal body weight per day intra- Four months following the transplant, he developed
venously for 4 days) without stem cell rescue in nine immune thrombocytopenia with hemolytic anemia
patients with severe refractory AIHA. The patients had (Evans’s syndrome). He received multiple courses of
not responded to a median of three (range, 1–7) other intravenous immunoglobulin, anti-Rh D immuno-
treatments. The median time to reach an absolute neu- globulin, a pulse of high-dose corticosteroids, and
trophil count of 500/μL or greater was 16 days (range, cyclosporine with some improvement of hemolytic
12–18 days). Six patients achieved complete remission, anemia, but no improvement of the thrombocytopenia.
and none relapsed after a median follow-up of Addition of vincristine resulted in long-term resolution
15 months (range, 4–29 months). Three patients of thrombocytopenia and anemia. No major toxicity
achieved and continued in partial remission. The was observed during treatment.
therapy was well tolerated.
Zupanska and coworkers11 reported that of 80
idiopathic and secondary AIHA patients originally INDICATIONS AND THERAPEUTIC REGIMENS
treated with prednisone, 43 needed additional treat-
ment with azathioprine or cyclophosphamide; 26 of On the basis of the preceding reports, immunosup-
these (60.5%) responded favorably. pressive drugs appear to be beneficial in some
Worlledge10 summarized her experience with the patients with WAIHA. Most clinicians suggest their
use of azathioprine as follows: 15 patients had been use after both corticosteroids and splenectomy have
treated for 3.5 months to 2.5 years; six patients had failed to produce an adequate remission, and if it is
responded well, two had responded partially, and six further demonstrated that an adequate remission is
had failed to respond. One patient presented with not possible using small doses of prednisone after
signs of Hodgkin’s disease after 2.5 years of treatment; splenectomy.165 Immunosuppressive drugs may be
one patient developed leukopenia; none became used instead of splenectomy, however, if contraindi-
thrombocytopenic. cations to surgery exist, but many physicians opt
Heisel and Ortega156 reported that seven children first for the use of other, less toxic measures.
with chronic AIHA who were steroid dependent had Several authors have recommended regimens for the
been treated with immunosuppressive drugs; only administration of immunosuppressive drugs to
one had achieved a complete remission. patients with AIHA.2,165-168 These regimens have been
Panceri and colleagues147 reported the case of a designed arbitrarily and are quite similar. Azathioprine
5-month-old boy with a life-threatening AIHA that in a dose of 1–2 mg/kg/day or cyclophosphamide
was unresponsive to therapy with steroids, high-dose (1.5–2 mg/kg/day) can be used to initiate cytotoxic
immunoglobulin, azathioprine, and splenectomy. drug therapy. If corticosteroids have been responsible
Despite these therapies, the patient’s condition wors- for an incomplete remission prior to the initiation of
ened, his hemoglobin level remained below 4 g/dL, therapy with immunosuppressive drugs, they should
and he required two to three blood transfusions daily. be continued until signs of hematologic improvement
Immunosuppression was increased by giving high- occur, at which time they should be reduced gradually
dose methylprednisolone (40 mg/kg/day) followed in dosage and discontinued if possible. Murphy and
by high-dose cyclophosphamide (10 mg/kg/day for LoBuglio166 recommend continuing immunosuppres-
10 days). The child showed a striking, sudden sive drug therapy at the initial dosage for 6 months in
improvement starting on the fifth day of high-dose those who respond, although this is evidently an
cyclophosphamide therapy, followed by complete empiric recommendation, and a shorter period of time
recovery. No major long-term complications were may be chosen. It is possible that, for adults, mainte-
observed. nance doses of 25 mg every other day or even twice
Silva and coworkers157 reported two patients with weekly might suffice.
severe AIHA who had failed multiple treatment modal- In patients who do not respond after 4 weeks of
ities and subsequently were treated with synchron- therapy, the alternative drug can be substituted, or the
ization of cyclophosphamide and plasma exchange. dose of the drug used initially can be increased by
418 Immune Hemolytic Anemias

increments of 25 mg/day every 2 weeks until a cytotoxic-treated patient. The same is true of a
response or limiting side effects occur. Particularly number of bacterial organisms: Listeria, Herellea,
important adverse effects are gastrointestinal intoler- Serratia, and the like.
ance and evidence of marrow depression as manifested To summarize, the most important immediate
by leukopenia, thrombocytopenia, or increasing problem for patients taking cytostatic drugs is infec-
anemia with reticulocytopenia. Additional adverse tion from a great variety of organisms, some common
effects of cyclophosphamide are hemorrhagic cystitis, and some exotic.
alopecia, and adverse effects on the reproductive Impaired Fertility. Immunosuppressive drugs can
system. Blood counts should be obtained weekly affect conception, pregnancy, fetal development, and
during the first month of therapy and for a similar lactation.169 An important risk of some of these
period of time after each dose increase. Thereafter, medications is that of sterilization.169 In treating non-
blood counts should be performed biweekly for several malignant disease of various types, there are substantial
months and, if they become stable, monthly thereafter. chances for long-term remission, periods when procre-
If a therapeutic response occurs while the patient is ation could be of the utmost concern to the patient.
being maintained at full dosage but hemolysis reap- These potential side effects represent serious problems,
pears during decrease of the cytotoxic agent, full which must be dealt with in discussing the use of the
doses should be reinstituted for several additional medications, especially with youthful patients.
months. If the relapse occurs after the patient has been Teratogenic Side Reactions. All drugs affecting
in remission for some time after all drugs have been nucleic acid and protein metabolism exhibit terato-
withdrawn, a trial of corticosteroids alone has a genicity in animals. The doses necessary to trigger this
chance of success and may be considered before pro- effect vary greatly from compound to compound, and
ceeding to additional cytotoxic therapy.166 with the same compound from species to species, con-
siderations that make it difficult to extrapolate con-
ADVERSE EFFECTS clusions from animals to humans. Men should be
apprised of the possibility of teratogenicity, and
Although some adverse effects of immunosuppres- women must be informed of the risks of congenital
sive drug therapy have already been mentioned, a malformations and must use effective methods of
more complete discussion of possible complications is contraception.169 Cytotoxic drugs may be used after
warranted. the first trimester to treat life-threatening disease,
Infection. There is little doubt that the major imme- although this would seem to rarely be necessary in
diate complication of cytotoxic drug therapy is infec- patients with AIHA.169
tion. It is also noteworthy that most patients, at the Neoplasia. One of the most difficult of all problems
dose levels recommended in the preceding para- related to cytotoxic agents is the potential threat of
graphs, seem to do well over extended periods neoplasia, perhaps first becoming apparent only
without apparent trouble from exogenous organisms. years after initial exposure to a drug.170-173 The
Specifically, pyogenic organisms do not seem to cause increased prevalence of neoplasia in the immunosup-
an unexpected amount of disease in these patients pressed patient after organ transplantation is an
provided that the granulocyte count is maintained at unequivocal fact. Admittedly, this information cannot
adequate levels. Granulocytopenia of less than 1000 necessarily be extended to cover those patients whose
polymorphonuclear leukocytes/μL is much more diseases require more modest doses of cytotoxic
likely to result in disseminated pyogenic infection that drugs.
is unresponsive to antibiotics. The explanation for the development of malignant
Even without the presence of leukopenia, several disease in patients treated with cytotoxic drugs is not
classes of agents can give trouble. Herpes zoster is very clear. The most commonly expressed view is that, in
common but usually heals uneventfully. Dissemination their immunosuppressed state, immune surveillance
of herpes zoster virus has been recognized, with death is defective and that mutant or perhaps virus-
from this organism; deaths from chickenpox pneumo- transformed cells that would be promptly destroyed
nia and systemic measles have also been reported. in a normal person are instead permitted to survive
Rarer viral agents apparently also produce major ill- and multiply.
nesses in the immunosuppressed patient.
Other organisms that cause “intracellular infections” NEWER AGENTS
are organisms to which cellular immunity appears to
be of more importance to the host’s defenses than is Cyclosporine. Cyclosporine has had a major effect
circulating antibody. Mycotic and mycobacterial infec- on human organ transplantation since its approval in
tions are in this group. 1983.146 It is a potent immunosuppressive drug,
Virtually all the fungi have been seen to disseminate reflecting its ability to block the transcription of
under cytotoxic therapy, usually with a fatal outcome. cytokine genes in activated T cells,174 and it has been
Infections with fungi that are ordinarily not of great used for therapy of a variety of autoimmune dis-
pathogenetic significance for man, such as Nocardia or eases.175 A number of reports attest to its effectiveness
Aspergillus, have been shown to be a hazard for the in patients with AIHA.
Management of Autoimmune Hemolytic Anemias 419

Emilia and coworkers176 used cyclosporine therapy Some authors have reported that cyclosporine,
for three patients with AIHA and one with Evans’s when used to treat patients with AIHA associated
syndrome. All patients had resistant, life-threatening with B-cell chronic lymphocytic leukemia, not only
hemolysis and were refractory to previous treatments. controled the hemolysis but also reduced the
The treatment protocol was as follows: Cyclosporine leukemic mass.184 In contrast, Emilia and colleagues185
was begun at an initial total dose of 5 mg/kg/day and treated a patient with B-cell chronic lymphocytic
administered twice daily for 6 days, after which the leukemia who had repeated episodes of AIHA that
dose was reduced to 3 mg/kg/day and continued were resistant to therapy, which included high-dose
with slight changes to maintain a serum level between prednisone, cyclophosphamide, antilymphocyte glo-
200 and 400 ng/mL. To increase cyclosporine blood bulin, and splenectomy. As a result of cyclosporine
concentration, low-dose prednisone (5 mg/day) was treatment, the patient’s hemoglobin level improved
added also.177 rapidly and was maintained at a normal level. After a
All three patients with AIHA achieved a complete 5-month decrease of the white blood count with a
response, and the patient with Evans’s syndrome reduction of lymph node size, however, the chronic
responded partially. All patients required continued lymphocytic leukemia progressed with an increasing
cyclosporine administration to maintain remission. WBC count and lymph node enlargement.
Adverse effects included a slight elevation of creatinine A number of reports of failure of cyclosporine
level but no persistent nephrotoxicity. Hypertension therapy in AIHA also exist.186-188
requiring drug therapy was always resolved with dose When cyclosporine is used to treat patients with
adjustments. The simultaneous administration of autoimmune disease, the potential for toxicity—
grapefruit juice enabled a 25% reduction of the drug particularly nephrotoxicity—during long-term use is
dose.178,179 An adverse effect of chlorambucil was seen a significant concern. Because patients are at risk for
in one patient, leading to a dramatic decrease of irreversible and perhaps progressive changes in renal
cyclosporine levels.180 The authors suggest that structure, one must consider the risk-benefit ratio
cyclosporine should be recommended for refractory carefully before deciding whether to treat them with
patients because it shows long-term efficacy and safety cyclosporine.175
and is able to maintain remission at low doses without Mycophenolate Mofetil. Mycophenolate mofetil
significant side effects. They further suggest that its use has a potent cytostatic effect on lymphocytes; this is
be considered before immunosuppressive chemothe- the principal mechanism by which the drug exerts
rapy to avoid myelotoxicity or mutagenic effects. immunosuppressive effects.189,190 The FDA has
Dundar and associates181 reported a patient with approved marketing of the drug for prevention of
Evans’s syndrome who was refractory to conventional rejection in patients with allogeneic renal trans-
and high-dose methylprednisolone treatment and to plants.191 It prolongs allograft survival and reverses
splenectomy. The patient experienced excellent graft rejection; it has also been used to treat rheuma-
benefit from cyclosporine; his hematological values toid arthritis and psoriasis. Nephrotoxicity and overt
were completely normal at the 12th month of therapy, hepatotoxicity have not been reported, but the drug
without any side effect from the drug. could be linked to bone marrow suppression and
Hershko and colleagues182 reported results of certain malignancies.191
cyclosporine treatment in two patients with AIHA Zimmer-Molsberger and coworkers192 used myco-
and one with Evans’s syndrome. All three were phenolate mofetil as second-line treatment for two
responsive initially to standard corticosteroid treat- patients with severe AIHA, both of whom had been
ment but relapsed despite continued therapy. All treated with 2-chlorodesoxyadenosine (2-CDA) for
improved significantly following the introduction of B-cell lymphocytic leukemia. Both patients had been
cyclosporine. Follow-up was a maximum of 8 months treated unsuccessfully for hemolysis with oral pred-
at the time of the report. nisolone therapy. One patient had an excellent response
Rackoff and Manno183 described the clinical course to mycophenolate mofetil, which resulted in the cessa-
of a 6-year-old child with severe Evans’s syndrome tion of blood transfusion support and a hemoglobin
who had experienced life-threatening episodes of level of greater than 12 g/dL for more than 32 weeks.
hemolysis despite the use of multiple therapeutic The other patient, who developed insulin-dependent
modalities. Cyclosporine was given at a dose of diabetes and varicella zoster infection while on pred-
10 mg/kg/day divided into two doses on alternate nisolone, was able to discontinue corticosteroid
days. The hemoglobin and platelet counts both therapy. His response was incomplete, but he did
improved, and it was possible to reduce the patient’s develop a decreased transfusion requirement.
prednisone dose from 2 mg/kg/day to as low as Howard and colleagues193 reported the use of
1 mg/kg every other day. With this regimen, the patient mycophenolate mofetil in four patients with AIHA
had less severe hemolytic anemia, was less thrombocy- and six with idiopathic thrombocytopenic purpura
topenic, and had fewer hospitalizations. At the time of (ITP). All four patients with AIHA showed a complete
the report, the patient had completed almost 2 years or good partial response to treatment.
of alternate-day cyclosporine and prednisone therapy, Rituximab. Rituximab (Rituxan) is a genetically engi-
with minimal toxic effects attributable to cyclosporine. neered chimeric murine/human monoclonal antibody
420 Immune Hemolytic Anemias

designed to target the CD20 antigen.194,195 CD20 expres- response ranged between 4 months and 2.7 years. The
sion is restricted to B-cell precursors and mature B cells authors stated that no unusual toxicity or complica-
and is not found on uncommitted hematopoietic pre- tions occurred as a result of the treatment.
cursor stem cells, plasma cells, dendritic cells, or other Grossi and colleagues203 treated five patients with
normal tissues. The drug is highly effective for in vivo autoimmune hematologic disorders with rituximab.
B-cell depletion, with B-lymphocytes becoming unde- Three patients had ITP, and two subjects had ITP
tectable in peripheral blood after a single infusion and plus AIHA (Evans’s syndrome). The patients were
recovering only 6 to 9 months after discontinuation of refractory to steroids, high-dose IgG, splenectomy,
treatment.196 The antibody was introduced for the treat- and immunosuppressive treatments that included
ment of B-cell lymphomas.196-199 Rituximab has also cytotoxic agents. Platelet counts increased and
been used in the treatment of immune thrombocytope- remained stable in one patient with ITP plus AIHA
nia200,201 and AIHA in an attempt to decrease the after 6 months, although further therapy was
number of platelet or anti-RBC antibody-producing B required. A decrease in hemoglobin was noted in
cells. Experience with the drug is accumulating rapidly; another patient with ITP plus AIHA who did not
many anecdotal experiences and a few rather small respond to therapy and subsequently expired. No
series of patients have been reported.201a improvement of platelet count was observed in the
The drug represents an appealing and promising remaining patients.
treatment modality for patients with the most severe Rai and coworkers204 reported treatment with a com-
and/or refractory forms of AIHA. In refractory or bination of rituximab, cyclophosphamide, and dexam-
chronic disease, the use of rituximab is attractive ethasone in eight patients with chronic lymphocytic
because it could reduce or avoid some side effects of leukemia who had AIHA. All subjects responded to
prolonged therapy with corticosteroids and other therapy, with one patient achieving a normal hemoglo-
immunosuppressive drugs. Serious adverse effects bin level. The median duration of response was
have also been reported, however (see the discussion reported as greater than 12 months, with one patient
that follows). experiencing a relapse after 11 months.
Clinical Effectiveness. Lee and associates202 Ahrens and associates205 described a 68-year-old
reported the results of rituximab treatment of six man with an 8-year history of AIHA refractory to
patients with AIHA. Three patients had cold agglu- treatment with prednisolone, azathioprine, cyclopho-
tinin syndrome, and three had WAIHA. Low-grade sphamide, mycophenolate-mofetil, and pulsed high-
lymphoma was diagnosed in three subjects. Prior dose dexamethasone. Rituximab was given once a
therapy consisted of steroids, chemotherapy, splenec- week for 4 weeks, with baseline therapy consisting of
tomy, and plasmapheresis, with disease duration prednisolone, 15 mg/day (Fig. 11-4). CD19-positive
ranging from 3 months to 20 years. Rituximab cells in the peripheral blood decreased from 4% to
(375 mg/m2) was administered once weekly for undetectable levels and remained below 0.05% of the
4 weeks. One patient received six infusions of ritux- lymphocytes. Although the DAT remained positive,
imab. Of five evaluable patients, all had complete hemolysis decreased during the 6 months of observa-
hematological responses as indicated by the hemoglo- tion after treatment, and his hemoglobin rose from 8.4
bin and hematocrit values. One subject was retreated to 12.3 g/dL. Although the reticulocyte count remained
with rituximab after 5 months, with continued elevated and the haptoglobin value remained low, the
response for an additional 10 months. The duration of patient became largely asymptomatic.

Prednisolone 15 mg/d

Previous therapy:
14 Pred Anti-CD20 (Rituximab)
Pred + Azt
13
Pred + Cyc FIGURE 11-4. Hemoglobin concentration after rituximab
12 Pred + MMF application (4 × 375 mg/m2) in a patient with refractory
Hemoglobin [g/dL]

Dexa autoimmune haemolytic anaemia. Pred, prednisolone; Azt,


11 azathioprine 2 mg/kg/d; Cyc, cyclophosphamide
10 1–3 mg/kg/d; MMF, mycophenolate mofetil 2 g/d; Dexa,
dexamethasone 40 mg (once). (From Ahrens N, Kingreen
9 D, Seltsam A, Salama A: Treatment of refractory autoim-
mune haemolytic anaemia with anti-CD20 (rituximab). Br J
8 Haematol 2001;114:244–245.)
7
6

–50 0 50 100 150


Days
Management of Autoimmune Hemolytic Anemias 421

Seipelt and colleagues206 reported a 65-year-old prine, vincristine, splenectomy, cyclosporine, plasma-
male with CLL and Evans’s syndrome. Therapy with pheresis, antithymocyte globulin, and transfusions.
cyclophosphamide, vincristine, and prednisone lead Subsequent treatment with two courses of rituximab,
to an improvement of hemolysis, but the patient each consisting of four doses of 375 mg/m2 at weekly
remained thrombocytopenic. Subsequently, lympho- intervals, led to complete remission, which was sus-
cytes increased to 135,000/μL, and cyclophosphamide tained for 19 months at the time of the report.
3 g/m2 was given without effect on the peripheral Seeliger and colleagues210 reported the clinical
lymphocyte count. At this stage, therapy with rituxi- course of a 6-year-old boy with refractory AIHA. Due
mab (375 mg/m2 four times weekly) was initiated. to failure of conventional immunosuppressive
The platelet count normalized within 1 week after the therapy, an autologous peripheral blood stem cell
first dose of rituxuimab, and the lymphocyte count transplantation was performed. He showed a partial
dropped within 2 months to 11,000/μL. response, with a reduced demand for RBC transfu-
Circulating CD19 and CD20 cells were no longer sions. Due to persistence of the hemolytic process,
detectable within 2 weeks of treatment, and B cells did however, he was started on rituximab therapy on the
not reappear in blood until 5 to 9 months after the last 40th day after transplantation. Following two doses of
treatment. Normal counts were then reached within rituximab, the patient improved rapidly and devel-
the following month in all patients. Immunoglobulin oped a sustained complete response. After 10 months,
concentrations in serum fell below normal values hemolysis recurred and again responded to rituximab
for age during the same period. Five patients were therapy without the necessity for RBC transfusions.
kept on prophylactic intravenous immunoglobulin Some 15 months after initial antibody treatment,
replacement for 9 to 10 months after the last rituximab however, the patient developed a second relapse,
infusion. Normal hemoglobin concentrations and which was now refractory to rituximab therapy,
reticulocyte counts of fewer than 100 × 109/L were although CD20+ B lymphocytes were cleared from the
achieved in all cases within 4 months; these levels peripheral blood. The authors concluded that ritux-
persisted until the last follow-up, 15–22 months after imab and autologous peripheral blood stem cell trans-
the start of rituximab. Coombs tests became negative, plantation are important, though not curative,
and associated autoimmune features resolved within elements in the treatment of patients with severe
12 weeks. AIHA who are refractory to conventional immuno-
Rituximab’s effectiveness was not limited to the suppressive therapy.
period of profound B-cell depletion, as no patient Further favorable results of rituximab therapy for
relapsed thereafter within a 5- to 14-month follow-up. warm antibody AIHA have been reported by a
The authors concluded that the optimal number of number of other investigators,211-213 including a report
rituximab infusions required to be effective still of one patient with mixed warm and cold AIHA.214
remains to be determined. They suggested that a Also see the section on cold agglutinin syndrome later
reduction to one or two injections—which would limit in this chapter.
the duration of the B-cell deficiency—would be worth Rituximab in Young Children. Quartier and col-
assessing. leagues215 reported the effectiveness of rituximab in
Perrotta and colleagues207 reported an 18-year-old six children with severe AIHA that was refractory to
girl with SLE and life-threatening AIHA that did not prednisone therapy that included pulsed intravenous
respond to steroids, intravenous immunoglobulin, or methylprednisolone or to combination therapy with
cyclosporin A. Rituximab was given weekly at 375 immunosuppressive drugs, and (in two cases) to
mg/m2 for two doses. The drug was well tolerated, splenectomy. Four rituximab infusions were given
and the patient experienced no adverse effects. Her intravenously at a dose of 375 mg/m2 once a week.
hemolytic disorder ameliorated markedly, with a pro- Patients three and five received eight additional infu-
gressive increase of hemoglobin levels, starting a few sions at the same dose over the course of 14 weeks.
days after therapy. The patient remained disease-free Results are illustrated in Figure 11-5.
7 months later. Ng and coworkers216 reported the case of an 8-
Chemnitz and coworkers208 reported a 63-year-old week-old infant with fulminant AIHA refractory to
female with chronic lymphocyte leukemia who conventional therapy. Massive hemolysis resulted in
developed AIHA that was unresponsive to cortico- cardiac decompensation and acute renal failure,
steroids and three cycles of chemotherapy with cyclo- which necessitated mechanical ventilation and peri-
phosphamide. She was treated subsequently with toneal dialysis. Rituximab halted progression of the
rituximab (375 mg/m2 intravenously for 4 weeks). The hemolytic process, but the patient died of acute viral
patient’s condition improved rapidly, transfusions pneumonia and disseminated fungal infection.
were no longer needed, and prednisone was reduced to Zecca and associates196 described the case of an 18-
10 mg per day. month-old child with immune-mediated pure red cell
McMahon and associates209 described a 16-year-old aplasia and AIHA refractory to first- and second-line
boy with AIHA who repeatedly relapsed after aggres- immunosuppressive therapy, who was successfully
sive therapy over a period of about 8 years. treated with rituximab. The authors used replacement
Treatment included corticosteroids, IVIG, azathio- therapy with intravenous immunoglobulins, as the
422 Immune Hemolytic Anemias

CD20 lymphocytes (×10 9/L)


4000
3500
3000
2500
2000
1500
1000
500
0

Reticulocyte counts (×10 9/L)


600
500
400
300
200
100
0

Hemoglobin concentration (g/L)


500
400

300
100
0

Prednisone dose (mg/kg per day)


3.5
3.0
2.5
2.0
1.5
1.0
0.5
0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21
Months after start of rituximab

Patient 1 Patient 2 Patient 3 Patient 4


Patient 5 Patient 6

FIGURE 11-5. Changes in biological variables and prednisone treatment during rituximab therapy. (From Quartier P, Brethon B, Philippet P,
Landman-Parker J, Le Deist F, Fischer A: Treatment of childhood autoimmune haemolytic anaemia with rituximab. Lancet 2001;358:1511–1513.)

reduction of B-cell numbers can be expected to reduce count was observed in patients with concomitant
serum levels of IgM and IgG, leading to a possible risk thrombocytopenia (Evans’s syndrome). Three respon-
of infectious complications. der patients had relapse 7, 8, and 10 months, respec-
Subsequently, Zecca and colleagues217 reported the tively, after rituximab infusion. All three children
results of the use of rituximab for the treatment of 15 received a second course of rituximab, again achiev-
patients with AIHA resistant to conventional treat- ing disease remission. The authors concluded that
ment. All patients had previously received two or rituximab is both safe and effective in reducing or
more courses of immunosuppressive therapy; two even abolishing hemolysis in children with AIHA and
patients had undergone splenectomy. After complet- that a sustained response can be achieved in the
ing treatment, all children received intravenous majority of cases.
immunoglobulin for 6 months. With a median follow- Hongeng and colleagues218 reported on a 37-month-
up of 13 months, 13 patients (87%) responded. Median old boy who had β-thalassemia major and who under-
hemoglobin levels increased from 7.7 g/dL to a went unrelated T-cell–nondepleted bone marrow
20-month post-treatment level of 11.8 g/dL; reticulo- transplantation. On day 180 after the transplantation,
cyte counts decreased; and an increase in platelet a diagnosis of AIHA was made, and it proved to be
Management of Autoimmune Hemolytic Anemias 423

refractory to corticosteroids and IVIG therapy. After from 1 to 13 weeks following rituximab exposure.
two doses of rituximab, hemolysis decreased and Patients experiencing a severe mucocutaneous reac-
remained so during 3 months of observation (hemo- tion should not receive any further infusions and
globin improved from 5 g/dL to 10 g/dL), and corti- should seek prompt medical evaluation.
coseroids were tapered and discontinued in 1 month Huhn and colleagues224 described a 65-year-old
after the second infusion. patient who, 10 hours after the first dose of rituximab,
Although these results in young children are encour- developed multiorgan failure with renal failure, ele-
aging, a note of caution was sounded by Matthews219 vation of serum potassium to 6.6 mM, acidosis, and,
because there have been scattered reports of oppor- finally, cardiac arrest.
tunistic infections following rituximab therapy for Since recovery of B-lymphocytes after administra-
pediatric patients. Pneumocystis carinii and varicella tion of rituximab has been described as starting after
pneumonia were reported by Motto and coworkers,220 6 to 9 months and being completed only after
and enteroviral meningoencephalitis associated with 12 months, a significant decline in IgM and IgG serum
particularly prolonged B-cell depletion was reported levels can be expected, and the possibility of infec-
by Quartier and associates.221 Matthews219 recom- tious complications is a concern.207,225 There have
mended longer-term study of the impact of rituximab- been several reports of such complications, and this
induced B-cell depletion with larger numbers of has led some physicians to give replacement therapy
patients to better define the risk-benefit ratio in the with intravenous immunoglobulin.207,217,226-228
treatment of autoimmune disorders. Rituximab has also been reported to cause severe
Adverse Effects. The toxicity of ribuximab in patients worsening of mild AIHA in a patient with B-cell
with non–Hodgkin’s lymphoma has been reviewed in CLL.229 The patient was a 64-year-old male who had
detail.222,223 Most adverse effects are mild to moderate progressive disease, for which he had received several
and can be managed effectively with premedication, courses of standard therapy. Subsequently, rituximab
supportive care, and adjusted infusion rates.223 was administered for 4 consecutive weeks. Before
Nevertheless, for the 36,000 patients treated with rituxi- rituximab administration, the patient had evidence of
mab since its product launch in 1998 through mid-1999, mild hemolysis (hemoglobin 11.9 g/dL), but just after
a total of 675 medically confirmed adverse reaction the fourth administration of the drug, the hemoglobin
reports were received worldwide, and 494 of these level dropped to 4.6 g/dL, the reticulocyte count was
reactions (73%) were classified as serious according to 23%, LDH and bilirubin levels were increased, and the
criteria defined by the International Congress for haptoglobin level was reduced. The DAT was found
Harmonization.222 The postmarketing surveillance positive due to IgG (3+) and complement (weak), and
database includes reports of 18 deaths that were judged the hemolysis responded quickly to prednisone
to be caused by infusion-related adverse reactions therapy. The authors suggested that massive destruc-
similar in type to those commonly reported during tion of CD20-positive cells by rituximab might create
early clinical trials. The infusion-related symptom an important liberation of cytokines (especially IL-6),
complex consists of fever and chills or rigors, which leading to the overexpression of antierythrocyte autore-
occur in the majority of patients during the first ritux- active plasmacytes (which were CD20-negative).
imab infusion. Other frequent infusion-related symp- Danazol. A possible efficacy of danazol, an attenu-
toms included nausea, urticaria, fatigue, headache, ated androgen, has been reported among patients
pruritus, bronchospasm, dyspnea, the sensation of with AIHA.230-237 Ahn231 has reported results of treat-
tongue or throat swelling, rhinitis, vomiting, hypoten- ment of 28 patients. Excellent or good responses
sion, flushing, and pain at disease sites. Serious car- were obtained in 77% of patients with idiopathic
diopulmonary infusion reactions culminating in death AIHA and in 60% with secondary disease. When the
have been reported to occur in approximately drug was discontinued after 1 year or more of
0.04%–0.07% of patients. The tumor lysis syndrome has therapy, lasting, unmaintained remissions of up to
been reported within 12–24 hours after the first anti- 5 years were often observed. The side effects of
body infusion and is estimated to occur in 0.04%–0.05% danazol are generally much less serious than those of
of patients. Major risk factors include high numbers of glucocortocoids, which can often be tapered or dis-
circulating malignant lymphoma cells, pulmonary continued completely.
infiltrates or lymphoma involvement, and prior cardio- Pignon and colleagues234 reported the results of
vascular disease. There are two reports of hemolytic treatment of 17 adult patients with WAIHA. For
anemia and one occurrence of transient aplastic anemia 10 patients, danazol (600–800 mg daily) was used,
following rituximab therapy. with prednisone (1 mg/kg/day) as first-line therapy.
In May 2001 the manufacturer distributed updated Among these patients, there were eight excellent
safety information, which indicated that there have responses and two failures. The mean follow-up
been 20 postmarketing reports of severe mucocuta- for responding patients was 21 months. A second
neous reactions associated with the use of rituximab group of patients included five who were initially
in an estimated 100,000 patients since product launch. treated with prednisone and who relapsed, and two
Eight of these cases resulted in fatal outcomes. The patients with refractory AIHA who had failed to
onset of the reaction in the reported cases has varied respond to various therapies, including splenectomy
424 Immune Hemolytic Anemias

and immunosuppressive therapy. In this group, an cluster on human chromosome 1q32 and share a
excellent response was observed in three of the five common 60–70 amino-acid short consensus repeat
patients who had relapsed after initial prednisone (SCR).239 An excellent review of complement has been
therapy. One partial response and one failure were published (also see Chapter 4).240,241
observed in the two refractory AIHA patients. A number of strategies for inhibiting complement
Manoharan232 reported that five patients with AIHA have been devised, including the therapeutic use of
responded to danazol therapy and that three had a soluble recombinant proteins derived from the RCA
complete response. family, the transfer of native RCA proteins from
Cervera and associates235 reported on three patients exogenously administered RBCs, and the overexpres-
with SLE and coexisting AIHA who were treated with sion of native complement regulators, which has been
danazol. The hemolysis was unresponsive to pred- achieved by culturing normal cells ex vivo followed
nisone, which was used at a dose of 1 mg/kg/day or by reimplantation.242 As the half-lives of the inhibitors
higher for at least 1 month. One of the patients had are short, the limited clinical trials that have been
had a prior splenectomy. Danazol was commenced at undertaken have been done in disease states in which
200 mg/day and was added to the corticosteroid complement activation is well demarcated, as in
therapy. Dosage was increased stepwise by 200 mg reperfusion injury of the allograft in lung transplanta-
daily every 4 weeks, with a maximum dose of tion. In the case of chronic diseases such as SLE and
1200 mg/day. When the hemolysis had resolved for at AIHA, the study and use of recombinant complement
least 1 month, treatment with corticosteroids was inhibitors is not yet a reality.239
tapered, and treatment with danazol continued. If Kirschfink243 pointed out that with increasing
patients had sustained remissions with danazol or evidence that complement activation significantly
experienced side effects, the doses were gradually contributes to the pathogenesis of a large number of
reduced to 200–400 mg/day. Two patients, including inflammatory diseases, strategies that interfere with
the patient with a prior splenectomy, had excellent its deleterious action have become a major focus in
responses lasting for 25 and 42 months, respectively, pharmacological research (Fig. 11-6). Endogenous
at the time of the report, and they were able to taper soluble complement inhibitors (C1 inhibitor, recombi-
glucocorticoid therapy significantly. The third patient nant soluble complement receptor 1, antibodies)
responded well but developed jaundice, hepato- blocking key proteins of the cascade reaction, neutral-
megaly, and splenomegaly, necessitating discontinua- izing the action of the complement-derived anaphyla-
tion of the drug after 41 months of treatment. After toxin C5a, or interfering with complement receptor 3
withdrawal of danazol, signs and symptoms of (CR3, CD18/11b)-mediated adhesion of inflammatory
hepatic disease disappeared, and the patient had a cells to the vascular endothelium have been tested
relapse of hemolytic anemia. The authors state that successfully in various animal models in recent years.
danazol was well tolerated by most patients, although Promising results consequently led to clinical trials.
undesirable effects severe enough to necessitate dis- Basta244 has suggested that the mechanism of the
continuation of the drug have been reported. These therapeutic effect of high-dose IVIG (see later discus-
include weight gain, dizziness, rash, pseudotumor sion) could be interference with the complement
cerebri, hepatic adenoma, cholestatic hepatitis, and system. This conclusion is based on the results
thrombocytopenia. obtained in animal models of complement-mediated
Chan and Sack236 reported on one patient with SLE pathology, in vitro complement assays, and studies on
and severe AIHA that did not respond to therapy with related human diseases.
corticosteroids, splenectomy, azathioprine, chloram- Yazdanbakhsh and colleagues245 assessed the ability
bucil, and intravenous immunoglobulin (IVIG). The of a human recombinant soluble form of complement
patient responded to danazol, with maintenance of receptor 1 (sCR1) to inhibit complement-mediated
her hemoglobin and reductions in her transfusion and RBC destruction in vitro and in vivo. Treatment with
corticosteroid requirements. sCR1 increased the survival of transfused human
There seems to have been a decrease in interest in group A RBCs by 50% in the circulation of mice with
danazol as therapy for AIHA, as indicated by the fact preexisting anti-A for 2 hours after transfusion,
that, with the exception of the study by Cervera and reduced intravascular hemolysis, and lowered the
associates,235 no articles emphasizing its use have levels of complement deposition (C3 and C4), but not
been published in the last decade. IgG or IgM, on the transfused cells by 100-fold. The
Complement Inhibitors. Complement is an impor- authors suggested that their data highlight a potential
tant effector system of host defense. It consists of more use of CR-1–based inhibitors for prevention of
than 35 proteins, which in their native state either cir- complement-dependent immune hemolysis.
culate as serum-soluble components of the blood or are Intravenous Immunoglobulin. The efficacy of IVIG
associated with cellular membranes.238 A focal point of treatment of autoimmune thrombocytopenia and
regulation of complement activity is at the level of neutropenia suggested that it might also be beneficial
C3/C5 convertases. Regulation occurs through the for treatment of AIHA. IVIG is frequently used as a
action of a number of plasma proteins, all of which are second-line therapy for AIHA for patients who do not
members of the complement activation (RCA) gene respond to corticosteroids. The choice of IVIG is in part
Management of Autoimmune Hemolytic Anemias 425

CAB-2
Classical pathway sCR1/sCR1-sLex Alternative pathway
Serine proteinase
C1 C3b
FIGURE 11-6. Complement as a target in C4 inhibitors
B
antiphlogistic therapy. Complement activa-
C2 P
tion, potentially leading to severe inflamma-
Coagulation/ D
tion, can be blocked by the administration of C1 inhibitor
either physiological regulatory proteins, kinin systems
blocking antibodies, receptor antagonists,
C3 Compstatin
RNA aptamers, serine proteinase inhibitors,
or intravenous application of high-dose

Inflammation
Anti-C5
immunoglobulins. (From Kirschfink M: C5 C5a C5aR
Targeting complement in therapy. Immunol
Rev 2001;180:177–189.)
Inhibition
Anti-C5a C5aR
C5b-9 antagonist

because of its relatively low incidence of adverse effects proportion to their relative concentration in plasma
compared with other treatment options. Its effective- (Fig. 11-8). Bleeker and colleagues252 reported experi-
ness is rather disappointing, however, and it is quite mental data supporting such a mechanism and con-
expensive. cluded that a single high dose of IVIG induces a
Mechanisms of Action. Numerous mechanisms of relatively small but long-lasting reduction of autoanti-
action have been postulated regarding the immuno- body levels by accelerated IgG clearance. This mecha-
regulatory effects of IVIG.246-248 Some of these are nism can fully explain, as the sole mechanism, the
listed in Table 11-11 and are illustrated in Figure 11-7. gradual decrease in autoantibody levels observed in
They include the following: several patient studies. In some clinical studies,
however, larger or more rapid effects have been
1. Antigen-specific suppression through antimicrobial observed that cannot be explained by accelerated
antibodies or antiautoantigen antibodies. clearance. Hence, IVIG also can reduce autoantibody
2. Immunomodulatory effects that could be mediated levels through mechanisms such as down-regulation
by inhibition of phagocytosis by blocking Fc recep- of antibody production or neutralization by anti-
tors, or by effects on B-cell activation and changes idiotypic antibodies.
in T-cell distribution and function, in part related to
anti–T-cell receptor antibodies. Also, modulation of TABLE 11-11. IMMUNOREGULATORY EFFECTS OF
the immune system by idiotypic antibodies could IMMUNE GLOBULIN
play a critical role in the establishment of long-term
remission for patients with autoimmune disor- Fc receptors
ders.249 Idiotypic antibodies can Blockade of Fc receptors on macrophages and effector cells
a. Neutralize an autoantibody, preventing the Induction of antibody-dependent cellular cytotoxicity
binding of the autoantibody to its antigen and Induction of inhibitory Fcγ receptor IIB
Inflammation
facilitating the clearance of the autoantibody Attenuation of complement-mediated damage
b. Downregulate the B-cell receptor for a specific Decrease in immune-complex-mediated inflammation
antigen, thereby decreasing the autoantibody Induction of anti-inflammatory cytokines
production Inhibition of activation of endothelial cells
c. Exert effects mediated through regulatory T cells Neutralization of microbial toxins
Reduction in corticosteroid requirements
3. Other effects of IVIG could be a membrane stabiliz- B cells and antibodies
ing and an immune complex solubilizing effect. Control of emergent bone marrow B-cell repertoires
4. Finally, inhibition of deposition of early comple- Negative signaling through Fcγ receptors
ment activation products (C4b, C3b) onto target Selective down-regulation and up-regulation of antibody production
Neutralization of circulating antoantibodies by anti-idiotypes
surfaces has been suggested as a mechanism of T cells
action.246 Regulation of the production of helper-T-cell cytokines
Neutralization of T-cell superantigens
Masson250 and Yu and Lennon251 have suggested Cell growth
Inhibition of lymphocyte proliferation
that acceleration of the rate of IgG catabolism is the Regulation of apoptosis
most plausible unifying explanation for the beneficial
action of high doses of exogenous IgG in antibody- From Kazatchkine MD, Kaveri SV: Immunomodulation of autoimmune and
inflammatory diseases with intravenous immune globulin. N Engl J Med
mediated autoimmune disorders. Such a process 2001;345:747–755. Copyright © 2001 Massachusetts Medical Society. All
would eliminate individual IgG molecules in direct rights reserved.
426 Immune Hemolytic Anemias

FIGURE 11-7. Immunomodulatory effects of immune globulin on B cells and T cells. Arrows indicate the sites targeted for the effect of immune
globulin. Immune globulin interferes with the selection of B-cell repertoires, down-regulates or up-regulates antibody production, neutralizes patho-
genic autoantibodies and T-cell superantigens, modulates the activation and function of the effector T cells and the production by CD4 T cells of
cytokines mediated by type 1 and type 2 helper T cells, and controls cell growth. (From Kazatchkine MD, Kaveri SV: Immunomodulation of autoim-
mune and inflammatory diseases with intravenous immune globulin. N Engl J Med 2001;345:747–755. Copyright © 2001 Massachusetts Medical
Society. All rights reserved.)

A novel mechanism for the effectiveness of IVIG in out that no previous publication included results in as
AIHA was suggested by Mueller-Eckhardt and many as ten cases, and only one study reported more
coworkers.253,254 These investigators suggested that than five. These authors conducted pilot studies at
IVIG administration causes coating of RBC by IgG and three institutions, enrolling a total of 37 patients, and
that such IgG-coated RBC are phagocytosed by the combined these results with a review of 36 cases
cells of the mononuclear phagocyte system (MPS). The reported in the literature. This remains the largest
consequent saturation of the MPS would result in a study of the effectiveness of IVIG in AIHA.
decrease in phagocytosis of antibody-coated platelets All patients reported by Flores and associates255
and would explain why IVIG is generally effective in had WAIHA. Their report reviewed results in a very
immune thrombocytopenia. In AIHA, however, the heterogenous group of patients. There were 62 adults
MPS capacity might already be exhausted by autoanti- and 11 children; 34 of the patients had idiopathic
body-coated RBC, thus explaining the relative ineffec- AIHA, and 26 patients had an associated chronic
tiveness of IVIG in treatment of AIHA. lymphocytic leukemia or lymphoma. Other patients
Clinical Results. Although IVIG is probably used had immunodeficiencies, AIDS, or connective tissue
frequently in the treatment of AIHA, published disease. All but four patients either had received
reports of its effectiveness or lack thereof are surpris- prior prednisone therapy or were receiving it concur-
ingly scanty. In 1993, Flores and associates255 pointed rently. Indeed, it was because most patients showed
IHA-11(401-458) 11/18/03 4:47 PM Page 427

Management of Autoimmune Hemolytic Anemias 427

FIGURE 11-8. Regulation of catabolism of IgG by FcRn, a specialized intracellular Fc receptor. (From Yu Z, Lennon VA: Mechanism of intravenous
immune globulin therapy in antibody-mediated autoimmune diseases. N Engl J Med 1999;340:227–228. Copyright © 1999 Massachusetts Medical
Society. All rights reserved.)

little or no response after 2 or more weeks of initial was the second strongest predictor of treatment
corticosteroid therapy that they received a trial of response.
IVIG therapy. Some patients had also received The authors point out that the response rate for IVIG
immunosuppressive agents or plasmapheresis or had compares unfavorably with the efÀcacy of other treat-
had a splenectomy. Patients in the published reports ment modalities for AIHA, including corticosteroids
received 0.4–2 g/kg/day for 2 to 5 days; most and splenectomy. They suggest that high-dose IVIG be
received 0.4–0.5 g/kg/day for 5 days. The authors’ recommended only as adjunctive therapy for selected
patients received either 0.5 g/kg/day for 5 days or cases, such as for children in whom the toxic effects of
1 g/kg/day for 5 to 7 days. chronic high-dose corticosteroids and immunosuppres-
The authors used liberal criteria for a response. sive agents could be especially frequent and severe. No
They deÀned a “Type I response” as an increase of mention was made of the expense of high-dose IVIG,
hemoglobin of 2 or more g/dL within 10 days of initi- which is considerable.
ation of treatment with IVIG, regardless of the Ànal Subsequently, Ratko and colleagues257,258 published
hemoglobin level. A “Type II response” was deÀned the results of a consensus conference developed by a
as fulÀllment of the criteria of a Type I response plus University Hospital Consortium Expert Panel, which
achievement of a peak hemoglobin value of 10 g/dL considered off-label uses of IVIG preparations avail-
or greater. Data regarding hemoglobin levels were not able in the United States. All English-language review
available for all patients in the literature. articles (n = 201) and original reports (n = 1904) were
Of the 62 evaluable cases, only nine (14.8%) had a evaluated, and extracted data were reviewed by an
Type II response (Table 11-12).255 Type I responses expert panel. At the time of publication of the detailed
occurred in 25 of 62 patients (40.3%) (all Type II res-
ponses also qualiÀed as Type I responses and were
included in the Type I response total). A signiÀcant
shortcoming of the report is the lack of follow-up infor- TABLE 11-12. RESPONSE OF AIHA CASES
mation. The authors’ only comment about the duration TO IVIG TREATMENT
of response is that “the hemoglobin remained elevated
for three or more weeks in most cases.” Overall response rate 29/73 = 39.7%
Response rate in literature cases 17/36 = 47.2%
Contrary to previous reports,256 higher doses of IVIG Response rate in combined pilot studies 12/37 = 32.4%
(5 g/kg) could not be shown to increase the response Response rate in pediatric cases 6/11 = 54.5%
rate. There was a strong correlation of a low pre- Type I response rate 25/62 = 40.3%a
treatment hemoglobin level with treatment response Type II response rate 9/61 = 14.8%
(Table 11-13).255 Thirteen patients had a pretreatment a Eleven of seventy three patients were literature cases with no specific data
hemoglobin level of 6 g/dL or lower, and 12 of these available. All Type II responses also qualify as Type I responses (see text for
(92%) showed a Type I response. Four of these definitions) and are included in the Type I response total.
From Flores G, Cunningham-Rundles C, Newland AC, Bussel JB: Efficacy of
13 patients were among the 9 in the entire study with intravenous immunoglobulin in the treatment of autoimmune hemolytic anemia:
a Type II response. The presence of hepatomegaly Results in 73 patients. Am J Hematol 1993;44:237–242.
428 Immune Hemolytic Anemias

stabilized, followed by a gradual improvement at 21


TABLE 11-13. RELATIONSHIP BETWEEN THE days after IVIG without steroids. Treatment with
PRETREATMENT HEMOGLOBIN LEVEL (EXPRESSED steroids resulted in faster and higher increments in
IN GRAMS PER DECILITER) AND RESPONSE TO IVIG hematocrit levels. Patients did not have any episodes
THERAPY IN PATIENTS WITH AIHA of AIHA from 6 months to as long as 4 years while
receiving IVIG at 3-week intervals. The patient with
Hemoglobin Type I Type II No
Level Response Response Response
IgM-mediated AIHA was not benefited by IVIG
therapy.
≤6.0 12/13 = 92.3% 4/13 = 30.8% 1/13 = 7.7% Leickly and Buckley261 reported reversal of AIHA
6.1–7.0 4/9 = 44.4% 1/9 = 11.1% 5/9 = 55.6% with IVIG treatment at a dose of 450 mg/kg/day for
7.1–8.0 2/10 = 20.0% 0/10 = 0 8/10 = 80.0% 5 days in a patient with common variable immuno-
8.1–9.0 4/12 = 33.3% 3/12 = 25.0% 8/12 = 66.7%
≥9.1 2/16 = 12.5% 1/16 = 6.3% 14/16 = 87.5%
deficiency. At the time of publication, the remission
had been maintained for 34 months by single doses of
From Flores G, Cunningham-Rundles C, Newland AC, Bussel JB: Efficacy of 100–200 mg/kg at 4-week intervals or whenever the
intravenous immunoglobulin in the treatment of autoimmune hemolytic anemia:
Results in 73 patients. Am J Hematol 1993;44:237–242.
hemoglobin level fell or the reticulocyte count
increased.
Most reports of IVIG indicate its use in conjunction
proceedings in 1999, the effectiveness of IVIG had with other therapy, especially corticosteroids. In con-
been evaluated in a total of 77 patients with AIHA.258 trast, Otheo and colleagues262 reported the successful
All 18 reports were either case studies or series or treatment of high-dose IVIG when used as a single
open, uncontrolled trials. All of the reports were sum- therapy in a 4-year-old child with AIHA. The patient’s
marized in tabular form, including information on the hemoglobin was 5.2 g/dL, hematocrit was 13.9%, retic-
number of patients, dosage, and the responses ulocytes were 20%, and platelets were 55,000/μL. The
observed. Patients received initial total IVIG doses DAT was positive with anti-IgG but not anticomple-
ranging from 2 g/kg to 7 g/kg and maintenance infu- ment antibody; the IAT was positive without clear
sions for as long as 4 years. A positive response, specificity. IVIG was begun at 800 mg/kg/day for
signified by a hemoglobin concentration increasing 5 days. Hemoglobin and platelet counts reached
into the normal range, occurred in 40 patients (52%). normal values rapidly, and the reticulocyte count was
In contrast to the report by Flores and colleagues,255 2.7% on the fifth week after IVIG. A maintenance dose
data were not supplied concerning the pretreatment of 800 mg/kg was given every 2 weeks for 8 months.
hemoglobin level. Most IVIG recipients had WAIHA There was no trace of IgG autoantibody at the fifth
refractory to other therapies, including corticos- month, and 18 months later, he still had normal hema-
teroids, cytotoxic drugs, and splenectomy. Although tological values.
some patients experienced sustained responses to Successful treatment of AIHA with IVIG has been
IVIG treatment, many required maintenance doses. reported in a patient with AIDS,263 in a patient with
No clinically significant adverse events related to refractory and life-threatening AIHA in a man with
IVIG infusion were reported in these studies. The the primary antiphospholipid syndrome,264 and in
report concludes by stating that it is difficult to evalu- four patients with thalassemia major who had a large
ate the clinical value of IVIG therapy for AIHA ade- increase in blood consumption following the develop-
quately, primarily because no data from randomized, ment of RBC autoantibodies.265
comparative trials are available. The routine use of A number of reports fail to show any significant
IVIG is therefore not recommended. It could have a effect of IVIG in AIHA. 253,254,266,267
role in treating patients with WAIHA that does not Adverse Effects. No clinically significant adverse
respond to corticosteroids or splenectomy, or in treat- events related to IVIG infusion in patients with AIHA
ing patients in whom corticosteroids or splenectomy were reported in the comprehensive reviews just
are contraindicated. cited.255,257,258 Adverse effects have been reported in
A further consensus panel was convened in 2000 to other publications, however.
evaluate and define the therapeutic areas in which The risk of transmitting infectious agents accompa-
IVIG has demonstrated effectiveness. AIHA was not nies the use of almost any blood-derived pharmaceu-
mentioned in the report of this panel.259 tical preparation. The steps used in preparing the
Selected Case Reports. Besa260 reported eight IVIG products available in the United States,
patients with WAIHA in association with a lympho- however, practically eliminate the risk of transmitting
proliferative disorder. Seven patients had chronic HIV, HBV, and HCV. Although an outbreak of
lymphocytic leukemia, and one had Hodgkin’s hepatitis C in 1994 was attributed to IVIG, the manu-
disease. An additional patient with non– facturing process has since been changed, and the
Hodgkin’s lymphoma had AIHA caused by an IgM Centers for Disease Control and Prevention (CDC)
cold antibody. IVIG therapy was given at a dose of states that IVIG products should now contain essen-
0.4 g/kg/day for five doses, followed by maintenance tially no risk of HCV transmission.258
therapy every 21–28 days if evidence of recurrence Adverse effects, which are generally mild, can occur
was noted. Hematocrit levels of patients with WAIHA in 1% to 10% of IVIG recipients.268,269 These reactions
Management of Autoimmune Hemolytic Anemias 429

may include back or chest pain or sensation of chest and deep venous thrombosis of the leg or arm. Most
heaviness, chills, fever, headache, malaise, myalgia, of these events developed during or immediately after
nausea, vomiting, or renal damage. Sherer and col- IVIG infusion. The pathogenesis of the thrombotic
leagues270 reported that 36% of patients had at least events has been attributed to IVIG-induced platelet
one adverse effect, but that these effects were usually activation and increases in plasma viscosity.283 There
mild and transient. Patients who develop adverse is a dose-dependent increase in plasma viscosity with
effects during the first treatment course may be at increasing plasma immunoglobulin concentration.
increased risk of adverse effects during the subse- Reinhart and Berchtold287 reported a more than four-
quent course. fold increase in plasma immunoglobulin concentra-
In one retrospective study, aseptic meningitis with tion after IVIG therapy, which was accompanied by an
severe headache was reported in about 11% of increase in plasma viscosity from a mean of 1.26 cP to
patients who received high-dose IVIG therapy for 1.54 cP (reference value < 1.40 cP). This effect lasted
various autoimmune diseases.271 This complication up to 5 days after treatment. Two studies have shown
was more frequent among patients with a history of an increase in the incidence of fatal hepatic veno-
migraine headaches and occurred regardless of the occlusive disease when IVIG is used prophylactically
product type or infusion rate. Most adverse effects are to prevent transplant-related infections.291,292
associated with rapid infusion and usually resolve Patients most at risk for thrombosis are the elderly;
with temporary discontinuation or reduction in infu- patients with a history of vascular disease, thrombo-
sion rate. sis, or acquired or inherited thrombophilic disorders;
Acute renal failure was recognized as an adverse and patients with prolonged periods of immobiliza-
effect of IVIG therapy in 1987.272 Since then, it has tion.283 For these patients, IVIG can be infused at a
emerged as an important complication, and there is a slower rate (8 to 12 hours), and high-dose infusion
plethora of relevant reports.272-282 As of 1998, the FDA (400 mg/kg to 1 g/kg/day) should be avoided. IVIG
had received a total of 120 reports of adverse renal should also be used with caution for patients who
events (acute renal failure and renal insufficiency) have high levels of serum monoclonal protein and
associated with high-dose IVIG therapy.281 On review concomitant hyperviscosity.
of these case reports, 90% of the patients had received Rizk and associates293 reported on a 23-year-old,
sucrose-containing IVIG, 56% had diabetes mellitus, 91-kg male with multifocal motor neuropathy who
and 26% had pretreatment renal impairment. All renal developed transfusion-related acute lung injury
adverse events occurred in the first 7 days following (TRALI) 6 hours after receiving 90 g of IVIG over the
administration of IVIG therapy. course of 3 hours. Prior to the IVIG infusion, he
Acute renal failure, including fatal cases, has been received hydrocortisone (60 mg), diphenhydramine
reported predominantly in association with higher (25 mg), and famotidine (20 mg), all given intra-
doses of preparations containing sucrose that were venously, and acetaminophen (650 mg), given orally.
used during consecutive days.273 This finding led to Four hours after the IVIG infusion, he became
recommendations to ensure that patients are ade- increasingly dyspneic and coughed up pink, frothy
quately hydrated, to consider the risk of sucrose- sputum. Chest x-ray revealed bilateral interstitial
containing preparations in patients with risk factors and alveolar infiltrates consistent with noncardio-
for renal insufficiency, to limit the rate at which genic pulmonary edema or adult respiratory distress
sucrose-containing preparations are infused, and to syndrome. He recovered spontaneously with only
monitor renal function in patients receiving IVIG.274 bed rest and nasal oxygen. TRALI is thought to be
Levy and Pusey280 studied the incidence of renal caused by infusion of a blood component or deriva-
impairment in an unselected cohort of patients receiv- tive containing granulocyte- or HLA antibody, a bio-
ing two different preparations of IVIG over the course logically active lipid, or a cytokine that activates
of 20 months. A total of 287 courses of IVIG were adherent, primed granulocytes resulting in damage
administered to 119 patients for a variety of indica- to pulmonary endothelium.293 Antibody to granulo-
tions. Eight patients showed deterioration in renal cytes was detected in the IVIG product by indirect
function (6.7%), and in two patients (1.7%), no renal granulocyte immunofluorescence assay, and the
recovery occurred. There were no significant differ- authors propose that transfusion of a large quantity
ences in patient characteristics, dose, or preparation of of granulocyte antibody precipitated TRALI in their
IVIG administered to those patients with or without patient.
changes in serum creatinine. There was no association IVIG has also been implicated in episodes of im-
between the amount of sucrose in the IVIG and the mune hemolysis (see Chapter 12).
development of renal failure. The researchers con-
cluded that IVIG is associated with renal impairment Plasma Exchange
which could be irreversible, with a maximum inci-
dence of 6.7%. Plasma exchange has been used in the therapy of a
Twelve cases of IVIG-related thrombosis have been wide variety of disorders, particularly those associ-
reported in the literature.283-290 Adverse events were ated with a well-documented or suspected immuno-
myocardial infarctions, strokes, spinal cord ischemia, logic pathogenesis.294-298 This therapy would seem
430 Immune Hemolytic Anemias

logical for IgM-mediated hemolytic anemias, as IgM carried out 10 days later, and he was started on aza-
resides exclusively within the plasma compartment thioprine at a dose of 2 mg/kg/day; prednisone was
and therefore can be expected to be removed also continued. After the second exchange, his hemo-
efficiently by the exchange process. IgG, which is dis- globin stabilized, and it was eventually found poss-
tributed almost equally between the intravascular and ible to withdraw both prednisone and azathioprine.
extravascular spaces, is less efficiently removed and The authors suggested that plasma exchange could be
quickly re-equilibrates to fill the void created by indicated for acute reversal of severe hemolysis while
removal of the patient’s plasma. Nevertheless, thera- other therapies are taking effect. In this way, plasma
peutic successes and failures have been reported for exchange has advantages over other therapies that
both warm and cold antibody AIHA. might take days or weeks to become effective.
Published results generally consist of case reports of A similar approach that combines plasma exchanges
small numbers of patients, and the results have been with immunosuppressive agents has been recom-
inconsistent. Further, concomitant therapy with ste- mended. Silva and coworkers157 reported treatment of
roids and immunosuppressive agents often obscures a patient with AIHA using a protocol of synchronized
the contribution of plasma exchange to the outcome. In plasma exchange and cyctotoxic drug therapy origin-
a number of cases, the AIHA seems to stabilize, but ally developed for treatment of patients with severe
other acutely ill patients have not improved. Finally, SLE.304 A 46-year-old female had recurrent episodes of
favorable effects are generally short-lived. WAIHA that was not responsive to a wide variety of
Branda and colleagues299 reported on a patient who therapies (splenectomy, corticosteroids, IVIG, vin-
developed WAIHA while convalescing from a viral cristine, azathioprine, danazol, cyclosporin, erythropoi-
illness. The patient was treated on one occasion only etin, and 10 plasma perfusions over staphylococcal
with 3000 mL plasma exchange followed by transfu- protein A). Combined therapy was begun with three
sions of 2 units of whole blood. The hemoglobin that one-volume plasma exchanges followed by intra-
had been rapidly falling stabilized, and an autoanti-e venous cyclophosphamide on the third day. Subse-
antibody, which had a titer of two before exchange, quently, cyclophosphamide and prednisone were given
became negative. The patient recovered from the orally in tapering doses. This regimen quickly pro-
hemolytic anemia without further therapy. duced remarkable improvement, and by day 51 after
Patten and Reuter300 reported benefit from plasma starting the combined therapy, the patient had no evi-
exchange in a 45-year-old woman with Evans’s syn- dence of AIHA. The authors suggest that the favorable
drome. Before plasma exchange, the patient’s hema- outcome might have been caused by the enhanced
tocrit level could not be raised above 10% despite cytotoxic effect of cyclophosphamide on proliferating
8 units of red cells administered over a 4-day period. lymphocytes participating in the antibody rebound
After plasma exchange, the hematocrit level rose to phenomena, a suppression of B lymphocytes with daily
29%, the platelet count rose from 14,500 to 222,000/mL, cyclophosphamide/prednisone therapy, and/or for-
and the transfusion requirements declined to only mation of anti-idiotypic antibodies. The relative
2 units of red cells over the next 37 days. The patient effectiveness of plasma exchanges and the immuno-
subsequently had a severe exacerbation of hemolysis suppressive drugs in the outcome of therapy with this
that did not respond to plasma exchange, however, and protocol is not clear.
she expired. Somewhat similarly, Hughes and Toogood305 used
Rosenfield and Jagathambal301 commented that plasma exchange as a prelude to the administration of
plasmapheresis has not been useful in their experi- high-dose IVIG. In so doing, they achieved a complete
ence for patients with WAIHA. remission in a patient with refractory AIHA.
Garelli and colleagues302 described a patient with a Silberstein and Berkman306 reported results in two
hyperacute hemolytic crisis treated by combined patients with WAIHA. One patient’s initial treatment
plasmapheresis and exchange transfusion. Immedi- consisted of corticosteroids and transfusions, to which
ately afterwards, the hemoglobin level rose from plasma exchange therapy was added shortly after
2.6 g/dL to 9.8 g/dL. The DAT became weakly posi- admission. The patient improved and subsequently
tive, and the hemolytic crisis subsided. Thereafter, the required intermittent treatment with corticosteroids.
clinical and laboratory picture stabilized. The second patient was treated with corticosteroids for
Bernstein and colleagues303 reported on a 17-year- 9 days prior to initiation of plasma exchanges.
old boy with fulminating acute hemolysis of 3 weeks’ Transfusion requirements decreased, but parameters
duration that was unresponsive to massive doses of indicating hemolysis showed only mild improvement.
corticosteroids (up to 10 mg/kg/day) and splenec- RBC survival studies revealed no improvement attrib-
tomy. He required 20 units of RBCs over a utable to the plasma exchanges.
14-day period to maintain a hemoglobin concentra- In 1993, McLeod and coworkers298 reviewed 17 cases
tion above 4 g/dL. A splenectomy was performed, but of WAIHA treated with plasma exchange.306-311 In a
there was a rapid fall in hemoglobin concentration in number of cases, plasma exchange seemed to stabilize
the first 4 days after surgery. Plasma exchange was the disease in patients who had entered a period of ful-
performed, with 8.5 liters of plasma removed and minant hemolysis; however, other acutely ill patients
replaced with 5% albumin. A second exchange was seemed not to have improved.
Management of Autoimmune Hemolytic Anemias 431

McConnell and colleagues312 described favorable RESULTS IN ANIMAL MODELS


results in a 34-month-old child (see Chapter 9).
McCarthy and associates313 reported the smallest infant In 1974, Morton and Siegel325 transferred murine SLE to
(7.5 kg) to receive intensive plasma exchange therapy irradiated BALB/C mice by transplanting whole
(52 procedures) as treatment for AIHA. Although the marrow from NZB mice. This adoptive transfer with
patient’s clinical course was prolonged and complicated marrow grafts was subsequently confirmed for many
by cytomegalovirus infection with spontaneous perfo- autoimmune diseases in animals, including murine
ration of the colon, his recovery was eventually com- models of SLE, the antiphospholipid syndrome, insulin-
plete. Because of his small size, calcium gluconate was dependent diabetes mellitus, experimental autoim-
added to replacement fluids, and calcium levels were mune encephalomyelitis, and adjuvant arthritis.326,327
monitored closely. The apheresis machine and tubing The same experimental autoimmune diseases that
were routinely primed with RBCs, and fresh-frozen have been transmitted by transplantation have been
plasma (FFP) was substituted for 5% albumin during cured by transplants from healthy animals.319 A puta-
the second half of all procedures to maintain adequate tive graft-versus-autoimmunity effect is supported by
levels of procoagulants. The patient had remained experiments showing that allogeneic chimerism
healthy for 2 years at the time of publication. achieved using a sublethal radiation conditioning
Shehata and coworkers314 performed an extensive regimen followed by allogeneic transplantation can
review of randomized controlled trials published prevent the onset of diabetes and even reverse pre-
between 1976 through 1999 in which therapeutic existing insulitis in nonobese diabetic mice, whereas
apheresis was used for numerous indications. AIHA the same radiation protocol without allogeneic human
was not mentioned in this review. In a summary of stem cells was insufficient.328 Both allogeneic and
current indication categories endorsed by the AABB autologous bone marrow transplants have been
and the American Society for Apheresis, plasma shown to be capable of preventing or treating autoim-
exchange for AIHA is listed as a category III indica- mune disease in experimental animal models.329
tion, meaning: “Therapeutic apheresis is not clearly Van Bekkum323 reviewed the experimental basis of
indicated based on insufficient evidence, conflicting hematopoietic stem cell transplantation for the treat-
results, or inability to document a favorable risk-to- ment of autoimmune diseases. He points out that the
benefit ratio. Applications in this category may repre- discovery that autologous bone marrow transplanta-
sent heroic or last-ditch efforts on behalf of a tion (BMT) is equally effective as allogeneic BMT in
patient.”315 inducing complete remissions in rats with experi-
mental autoimmune diseases cleared the way for
clinical application. Actually, the experiments with
Hematopoietic Stem Cell Transplantation syngeneic BMT were included because of the (mis-
The majority of autoimmune diseases are controlled taken) assumption that they would serve as negative
more or less satisfactorily by conventional therapeu- controls.
tic manipulation of the immune system, but there is a
hard core of refractory, relapsing, treatment-resistant RESOLUTION OF AIHA AFTER ALLOGENEIC STEM
autoimmune diseases for which the term malignant CELL TRANSPLANTATION
autoimmunity has appropriately been proposed.316,317
The concept of using intense immunosuppression fol- A 5-year-old boy affected from infancy by relapsing,
lowed by allogeneic or autologous human stem cells life-threatening Evans’s syndrome was transplanted
to treat autoimmune diseases is based on encourag- successfully with HLA-identical sibling cord blood.330
ing results in experimental animals, serendipitous There was total disappearance of autoantibodies, but
cases of patients with both autoimmune disease and the patient died of acute liver failure 9 months after
malignancies who were allotransplanted for the transplantation. Another patient with Evans’s syn-
latter, and phase I/II trials in various disease drome developed 100% donor hematopoiesis, no graft-
states.318-320 Van Bekkum323 reported that more than vs.-host disease (GVHD), and a complete hematologic
500 patients have been treated with autologous stem and immunologic remission, which had persisted for
cells for severe refractory autoimmune diseases. 5 months at the time of reporting.319
Indeed, Burt and colleagues324 reported that, by per- De Stefano and coworkers331 reported on a patient
centage of transplantations performed, autoimmune with thalassemia intermedia and immune-mediated
diseases are the most rapidly expanding indication hemolytic anemia who relapsed 7 weeks after an
for stem cell transplantation. These authors con- autologous lymphocyte-depleted peripheral blood
cluded, however, that further improvements in the stem cell transplant. A complete remission, which had
efficacy and safety of both autologous and allogeneic lasted 18 months at the time of publication, was
stem cell transplantation procedures need to be obtained with allogeneic bone marrow transplanta-
developed, and larger cohorts of patients need to be tion from an HLA-matched, unrelated donor. The
studied to assess the full benefits of stem cell trans- authors suggested that a graft-versus-autoimmunity
plantation as a most promising new armamentarium effect could have been important in the eradication of
for the treatment of autoimmune diseases. the patient’s autoaggressive lymphocytes.
432 Immune Hemolytic Anemias

Oyama and colleagues331a reported on a 28-year-old It has been postulated that if immunosuppressive
male with Evans’s syndrome that was refractory to regimens can eliminate or effectively reduce the level
multiple interventions. He developed life-threatening of autoreactive T and B cells, then regeneration of de
complications and was treated with an allogeneic novo immunity even in the autologous setting could
hematopoietic cell transplant from his HLA-matched bypass the initial breakdown of self-tolerance and
sister. This resulted in complete clinical and serologic ensure prolonged disease remission.334
remission of the immune-mediated hemolytic anemia Although autologous transplants have been per-
and thrombocytopenia, which had persisted for more formed mainly for neurologic conditions such as multi-
than 30 months at the time of the report. The clinical ple sclerosis335 and for rheumatologic conditions,336 a
course after transplantation was complicated, how- smaller group of patients with refractory hematological
ever, by drug-related thrombotic thrombocytopeic autoimmune diseases also have undergone the proce-
purpura, chronic extensive GVHD, and immune sup- dure.319,337,338 Paillard and colleagues339 reported on a
pression-related opportunistic infections. case of a child with severe AIHA who did not respond
Marmont and associates332 reported on a 21-year- to conventional treatments but was cured with an
old male with refractory Evans’s syndrome (predom- autologous peripheral blood CD34+ cell transplant-
inantly thrombocytopenic) who was treated with an ation. No further RBC transfusions were required
allogeneic reduced-intensity bone marrow transplant beyond day 16 after the autograft. At 20 months after
from his HLA-identical sister. He had an initial dra- autograft, the patient was in complete hematological
matic platelet peak, but while still evidencing mixed remission.
chimerism he again became progressively thrombo- SLE, a disorder frequently complicated by immune
cytopenic. He finally remitted following five donor cytopenias, is becoming a major target for autologous
lymphocyte infusions and remained in complete clin- transplants.319,340,341 Musso and coworkers341 reported
ical and biological remission for 2 years after trans- on a 19-year-old female with a 6-year history of SLE
plantation. The authors state that evidence is with secondary antiphospholipid syndrome who later
accumulating that a graft-vs.-autoimmunity effect developed refractory Evans’s syndrome. She was
exists, consisting most probably in the substitution of treated with an autologous hematopoietic cell trans-
normal T, B, and lymphoid progenitor cells for the plant, and 8 months later she had normal blood counts,
autoimmune clones of the patient’s immune system. although with persistent low-titer direct antiglobulin
A role for graft-vs.-autoimmunity was also demon- and antinuclear antibody tests. Anti-double stranded
strated by Hinterberger and coworkers,333 who DNA, lupus anticoagulant tests, and anticardiolipin
reviewed reports of hematopoietic cell transplants for tests were negative.
autoimmune diseases published between June 1977 In one patient with SLE and non–Hodgkin’s lym-
and September 2001. They found that freedom of phoma, the lymphoma did not relapse, but autoim-
relapse was superior after allogeneic transplantation mune thrombocytopenic purpura supervened. The
compared with autologous transplants (P = 0.0002). autoimmune disease thus appeared more refractory
These data suggest that a graft-vs.-autoimmunity than the neoplasia.342 A similar sequence occurred in
effect after allogeneic hematopoietic cell transplanta- the case of a patient with Sjögren’s syndrome and
tion mediates elimination of autoimmunity. lymphoma; complete remission of the lymphoma
occurred, but thrombocytopenia and vasculitis
recurred 2 months after transplantation.343
AUTOLOGOUS TRANSPLANTS FOR
AUTOIMMUNE DISEASE
Intense Immunosuppression without
Although one hesitates to use the oxymoron “autolo- Human Stem Cell Rescue for Treatment
gous transplant,” it is a commonly used term in the
literature of transplantation and refers to high-dose
of Autoimmune Disease
marrow ablative therapy followed by rescue with A novel approach to circumventing the problem of re-
autologous hematopoietic stem cells. Such “trans- infusing autoreactive lymphocytes is to give an
plants” from marrow or from peripheral blood are immunoablative regimen that spares early hemato-
much more commonly used than allogeneic hemato- poietic precursors, obviating the need for an auto-
poietic cell transplants to treat autoimmune disease graft.344-347 Brodsky and associates344 found that
because of the greater safety of autologous proce- immunoablative doses of cyclophosphamide, without
dures, although transplant-related mortality has been stem cell rescue, can induce durable complete remis-
higher (8.6%) than initially anticipated (1–3%).334 sions (median follow-up >10 years) in the majority of
Some centers, however, have performed autologous patients with severe aplastic anemia. This approach has
transplantation on more than 70 autoimmune patients subsequently been extended to a spectrum of severe
with no transplant-related mortality.334 Possible autoimmune diseases including Felty’s syndrome,
reasons for center differences include the diseases autoimmune thrombocytopenic purpura, Evans’s syn-
transplanted, selection or exclusion criteria, and inten- drome, SLE, chronic inflammatory demyelinating poly-
sity of immune-suppressive preparative regimens. neuropathy,346,348,349 and paraneoplastic pemphigus.347
Management of Autoimmune Hemolytic Anemias 433

High-dose cyclophosphamide was well tolerated; Russell355 reported a 2.5-month-old infant with AIHA;
median times to neutrophil count of 500/μL and the infant was unresponsive to splenectomy or corti-
platelet transfusion independence were 17 and 16 days, costeroids, but thymectomy induced hematologic
respectively. Four patients (two with rheumatoid remission. The antiglobulin test was still positive
arthritis/Felty’s syndrome, one with SLE, and a patient 8 weeks after surgery, but it reverted to negative
with chronic demyelinating polyneuropathy) were in 3 weeks later. Cure of AIHA in an infant was also
continuous complete remission for 3 to 21 months after reported after thymectomy by Karaklis and associ-
treatment at the time of the 1999 report of Brodsky and ates.357 Subsequently, cases in which the operation pro-
Smith.346 These findings suggest that autologous duced benefit were reported by Hancock,356 Dacie and
human stem cells have mainly a rescue effect.348 Worlledge,94 and Hirooka and coworkers.359 No benefit
Further clinical trials are clearly indicated. was recorded in patients described by Oski and
Abelson358 and Johnson and Abildgaard.154
More recently, Rennenberg and colleagues360 repor-
FUTURE DIRECTIONS FOR INTENSE
ted on a case of thymoma-associated AIHA treated suc-
IMMUNOSUPPRESSION THERAPY
cessfully with a short-term course of prednisone and
The crux of the matter is whether intense immuno- subsequent thymectomy. The patient patient’s initial
suppression followed by rescue with human stem hemoglobin level was 3.7 g/dL, and reticulocytes were
cells is indeed capable of ensuring tolerance, or, in 38%. After treatment with prednisone, the hemoglobin
other words, of eradicating autoimmunity. This ambi- improved to 6.3 g/dL, and reticulocytes decreased to
tious goal has been achieved experimentally, but in 9%; in addition, there was a rapid decline in serum
clinical settings, reprogramming of the immune bilirubin and LDH levels, and the serum haptoglobin
system is still elusive.319 Early studies have involved level became normal. After the thymectomy, the hemo-
patients who had severe disease manifestations and globin improved to a level of about 8 g/dL, reticulo-
have been marginal candidates for high-dose condi- cytes decreased to approximately 2%, and signs of
tioning regimens. Whether the treatment would be hemolysis did not occur.
more tolerable and even more effective if moved to an Rennenberg and colleagues360 also reviewed the
earlier state of the disease remains the subject for literature and summarized an additional 15 cases
future study. Other issues to be explored are the need (Table 11-15).361-372 In seven of the 14 patients for whom
for removal of immune cells through CD34 selection pathological data were available, the thymoma was
and determining which conditioning regimen is best. reported as malignant, and in 5 of 14 patients (35%),
The introduction of nonmyeloablative, reduced- signs of pure red cell aplasia were also present, sug-
intensity conditioning regimens could reduce trans- gesting a common pathogenetic mechanism. The
plant-related complications and allow the more authors pointed out that most patients with thymoma-
widespread use of human stem cell transplants.350-354 associated AIHA were treated with corticosteroids, and
Even if stem cell transplantation has failed to this led to the disappearance of the hemolysis in 75% of
produce clinical results comparable to the results the patients treated. Thus, the role of the thymectomy
achieved in animal models, some significant benefits in causing remission is rather uncertain.
have been achieved, and future benefits are likely.
COLD AGGLUTININ SYNDROME
Thymectomy
Before 1980, thymectomy was performed on a number
Avoidance of Cold
of occasions as therapy for WAIHA, with variable Therapy for cold agglutinin syndrome (CAS) is often
results (Table 11-14).1,94,154,355-359 In 1963, Wilmers and unsatisfactory. It is fortunate that the disorder is

TABLE 11-14. THYMECTOMY IN AUTOIMMUNE HEMOLYTIC ANEMIA

Authors (Year) Age Course Reference Number

Wilmers and Russel (1963) 2.5 mo Cured 355


Hancock (1963) infant Successful 356
Karaklis et al. (1964) infant Cured 357
Oski and Abelson (1965) 6 wk No benefit 358
Dacie and Worlledge (1969) 7 mo Cured 94
Hirooka (1970) 8 yr Marked clinical improvement 359
Johnson and Abildgaard (1976) 7 mo No benefit 154
Pirofsky (1969)* 59 yr No benefit 12

* Associated with thymoma and erythrocytic aplasia.


434
TABLE 11-15. CHARACTERISTICS OF 16 PATIENTS WITH THYMOMA-ASSOCIATED DAT-POSITIVE
AUTOIMMUNE HAEMOLYTIC ANAEMIA

Treatment Follow-up

Author Sex/Age Interval Thymoma/AIHA Thymectomy Steroids Type Thymoma Reticulocytes (%) Haemolysis Coombs

Albahary (361) M/74 7 yr No No – 21 Recovery –


Arntzenius (362) F/82 Simultaneous Yes No Malignant lymphoid 12 Recovery Positive
and epithelial
Dreyfus (363) F/56 13 yr Noa Yes Spindle cell type 0 Recoveryb Negative
Fisher (364) M/31 3 yr Noa Yes Malignant 11.5 Recovery Positive
Halperin (365) M/62 Simultaneous Yesc Yes Malignant 23 Recovery Weakly +
Hennemann (365a) F/70 7 yr Nod Yes Malignant 11.3 Recovery Positive
Janbon (366) F/65 Simultaneous No Yes Malignant – No recoverye –
Mongan (367) F/63 7 yr Noa Yes Lymphoid 30.4 – Negative
Pirofsky (368) M/59 –10 mo Yes Yes Spindle cell type 0 No recovery Positive
Pirofsky (368) M/75 –34 mo No Yes Spindle cell type 1.4 Recovery –
Pirofsky (368) F/66 –26 mo No Yes – 6.8 Recovery Positive
Ross (369) F/44 3 yr Yes No Spindle cell type 0.1 Recoveryf Negative
Rubinstein (370) M/51 Simultaneous Yesc Yes Malignant lymphoid 20 Recovery Weakly +
and epithelial
Taniguchi (371) M/58 5 yr Noa Nog Epithelial 0 Recovery Negative
Tiber (372) M/48 >10 yr No Yesh Malignant 0.1 Recovery Negative
Case F/50 Simultaneousc Yesc Yes Spindle cell type 38 Recovery Weakly +
a Previous thymectomy.
b Recovery after a great variety of treatment modalities.
c Thymectomy was performed after steroid-induced recovery of haemolysis.
d Previous irradiation of the thymus.
e Death within one month.
f After splenectomy.
g The first episode of haemolysis subsided spontaneously, the second episode was successfully treated with steroids.
h Steroid-based chemotherapy. Data not available or not evaluable.
From Rennenberg RJ, Pauwels P, Vlasveld LT: A case of thymoma-associated autoimmune haemolytic anaemia. Neth J Med 1997;50:110–114.
Management of Autoimmune Hemolytic Anemias 435

frequently not severe and, instead, results in a chronic exclude the presence of warm hemolysins active
mild hemolytic anemia with a hemoglobin level of 9 to against enzyme-reated red cells, which are present in
12 g/dL. Such patients require no therapy other than about 10% of the patients who have WAIHA and
avoidance of cold (Fig. 11-9).6,373 If patients go out in have C3 (but not IgG) on their RBCs. Prednisone
cold weather, they must clothe themselves well, using administration at a dose of 100 mg daily on two
earmuffs, warm socks, and warm mittens. These meas- occasions to the first patient produced a “remission”
ures prevent the development of acute hemolytic crises, characterized by a rise in hemoglobin of more than
but even with strict avoidance of cold, some hemolysis 2 g/dL; in the second patient, similar doses of pred-
usually persists. Patients with moderate degrees of nisone caused a more impressive increase in the
anemia should usually be urged to learn to tolerate the hemoglobin level. The first patient required “at
symptoms rather than embark on therapeutic trials that least” 20 mg of prednisone, however, and the second
have greater potential risk than probable benefit. patient required 40 to 60 mg of prednisone to main-
tain a hemoglobin level of 10 g/dL.
Corticosteroid Therapy The high maintenance doses of prednisone required
to maintain a hemoglobin level of greater than
Corticosteroids are less effective for CAS than for 10 g/dL even in these “responsive” patients illustrates
warm antibody syndromes.6 Lack of success of corti- a major problem. Pirofsky,12 in a retrospective review
costeroid therapy in CAS has been reported by of his own series of patients, has emphasized strongly
Pisciotta,374 Firkin and coworkers,375 Dausset and that the maintenance of a partial remission in patients
Colombani,62 and Dacie.6 Some case reports, however, with AIHA by prolonged administration of pred-
do indicate that corticosteroids can be effective, often nisone at dose levels over 15 mg per day results in
for patients with atypical clinical and serological “disastrous side effects.” The tendency to develop
findings. such side effects is often underestimated by physi-
Schrieber and colleagues376 reported two patients cians. In CAS, educating the patient to tolerate a lower
who had a modest response to high doses of cortico- than normal hemoglobin level is ultimately likely to
steroids. The patients were diagnosed as having CAS be preferable to chronic prednisone therapy. Patients
with atypical serologic reactions. In each case, the with a hemoglobin level of 7 to 10 g/dL are often
DAT was strongly positive with anti-C3 and negative capable of tolerating the symptoms of anemia, and
with anti-IgG. The patients had only a modest they should be strongly urged to do so rather than be
elevation of the cold agglutinin titer at 4°C, but treated with high doses of prednisone (see Case
the antibodies reacted up to 37°C. Such serologic Report, Chapter 5). If patients are more anemic, have
findings do, indeed, occur in a number of patients intolerable symptoms, and are refractory to cytotoxic
with CAS (see Chapter 5). The authors did not drug therapy (to be described later), a program of

550 26.6

500 21.1

Mean monthly minimal temperature (°C)


Lactate dehydrogenase
Temperature
Lactate dehydrogenase (U/mL)

450 15.5

400 10.0

350 4.4

300 –1.1

250 –6.6

200 –12.2

150 –17.8
10 11 12 1 2 3 4 5 6 7 8 9 10 11 12 1 2 3 4 5 6 7 8 9 10 11 12

1992 1993 1994


FIGURE 11-9. A 61-year-old man with cold agglutinin syndrome worked outdoors in the mid-west area of the United States. The severity of his
hemolysis, as indicated by LDH values, was related to the ambient temperature. (From Lyckholm LJ, Edmond MB: Images in clinical medicine:
Seasonal hemolysis due to cold-agglutinin syndrome. N Engl J Med 1996;334:437.)
436 Immune Hemolytic Anemias

chronic transfusions is probably the optimal means of Immunosuppressive Drugs


management. In most instances, prednisone in accept-
able doses (15 mg per day for maintenance therapy) Very little has been added to the recent medical
offers no significant benefit. literature regarding immunosuppressive drugs for
Silberstein and coworkers377 studied patients who management of patients with CAS. Indeed, in the
had hemolysis associated with serum cold agglu- 1992 review by Dacie,6 the most recent of the refer-
tinins that were not inactivated or were incom- ences cited was published in 1982. Nevertheless, the
pletely inactivated with the IgM-reducing agent older literature contains some valuable and detailed
dithiothreitol. They determined the immunoglobu- information.
lin isotypes of the cold agglutinins in five patients.
In two patients, the antibodies were predominantly CYCLOSPHOSPHAMIDE AND CHLORAMBUCIL
IgG with smaller amounts of IgM or IgA; in two
other patients, IgM predominated, with lesser Cytotoxic drugs have resulted in favorable responses
amounts of IgG; one patient had an IgG cold agglu- in a minority of patients with CAS. Schubothe381
tinin only. These patients were atypical in their suc- described the use of cyclophosphamide in four
cessful therapeutic response to daily glucocorticoid patients. In two patients, the cold agglutinin titer
therapy and (in the case of two patients) splenec- decreased, but no clinical details are given except that
tomy. That patients with IgG cold agglutinins might “neither patient showed the picture of chronic cold
be expected to respond to glucocorticoids and agglutinin syndrome.”
splenectomy in a manner similar to patients with Worlledge and coworkers382 reported in 1968 on the
WAIHA is also suggested by the report of Dellagi use of chlorambucil given orally at a daily dose of
and colleagues.378 Even in some of the patients 2–10 mg to nine CAS patients for periods of between
reported by Silberstein and coworkers,377 however, 4 and 12 months. Three of the patients responded
the response to corticosteroids was unimpressive. temporarily, two patients were judged to have
Patient 1 was treated not only with corticosteroids responded partially, and three were not helped. The
but also with cyclosphosphamide, three plasma- cold aggglutinin titer and the serum IgM levels fell in
phereses, and multiple transfusions. He required patients who responded. Several patients experienced
prolonged treatment with glucocorticoids and unwanted side effects that included pancytopenia,
cyclophosphamide until a splenectomy was per- lymphopenia, reticulocytopenina, and non–Hodgkin’s
formed. Patient 2 required 30–40 mg per day to lymphoma. In 1982, Worlledge10 reported that of the
produce a 1–2 g/dL increase in hemoglobin level. 11 patients she had by then treated with chlorambucil,
Patient 3 had 2 weeks of therapy with prednisone, 5 had failed to respond.
after which a splenectomy was performed, thereby Hippe and colleagues383 reported the use of chlor-
making the effectiveness of the corticosteroids ambucil for four patients with CAS. Some of the
difficult to evaluate. Patient 5 had AIHA of only results are indicated in Table 11-16.383 All four patients
2 days’ duration and was treated with transfusions, presented with acrocyanosis and hemoglobinuria,
plasma exchange, and prednisone therapy. He and patient 4 also had gangrene of the fingertips.
improved after 2 weeks of therapy, after which pred- After therapy, none of the patients had acrocyanosis
nisone was tapered. The patient could have had an or hemoglobinuria, and two patients were able to
acute, self-limited episode of hemolysis, for which work outdoors in winter (in Denmark). Dosage sched-
prednisone provided little benefit. ules varied, and the patients were treated intermit-
Other case reports also describe a response to cor- tently or continuously. Effects were most pronounced
ticosteroid therapy in cold antibody AIHA. Nanan during the first 6 months, but for two patients,
and associates379 treated a 12-year-old boy suffering essentially normal values of IgM were obtained only
from severe low-titer CAS with methylprednisolone after treatment for periods of 1 year and 3 years,
at a dose of 20 mg/kg for 3 consecutive days. The respectively.
patient’s symptoms improved within 12 hours of the Olesen384 described one patient in whom therapy
first steroid infusion, and hemolysis was halted. with chlorambucil caused a reduction in the cold
Lahav and colleagues380 reported an unusual case of agglutinin titer from 128,000 to 32,000 and a decrease
idiopathic CAS that responded well to cortico- in thermal amplitude by 3°C. There also was a subjec-
steroids. The titer of the cold antibody was only 32 at tive improvement in the intensity of the patient’s acro-
4°C, and it reacted to a titer of 8 at 37°C; the antibody cyanosis. Her hemoglobin value, however, did not
reacted with both I and i RBCs. The patient change during one course of therapy, and during a
responded to prednisone, 60 mg per day, with second course, it dropped from 12.0 g/dL to 9.5 g/dL.
improvement in the hemoglobin level from 8.6 g/dL Evans and coworkers385 described a patient with a
to 11 g/dL. She was followed for 6 years, and it was chronic CAS of varying severity over a 20-year
found that she needed 20 mg of prednisone daily to period. Chlorambucil therapy (6 mg per day for
prevent the recurrence of hemolysis. No signs of a 3 months) resulted in a decrease in cold agglutinin
lymphoproliferative disorder or other underlying titer from 64,000 to 2000, an increase in hematocrit
disease developed during this period. from 29% to normal levels, a decrease in reticulocytes
Management of Autoimmune Hemolytic Anemias 437

TABLE 11-16. LABORATORY DATA BEFORE AND AFTER TREATMENT OF COLD AGGLUTININ SYNDROME
WITH CHLORAMBUCIL

Dates of Serum IgM Concentration Cold Agglutinin Titer in Decrease in


Patient (Age) Laboratory Tests (N = 0.11 – 1.23 g/L) Percentage of Initial Titer Thermal Amplitude (°C) Hemoglobin (g/dL)

1 (78) 1962 7.2 100 – 11.5


1968 1.0 17 3.3 13.5
2 (65) 1966 6.6 100 – 9.0
1968 1.8 37 2.2 10.0
3 (62) 1967 8.2 100 – 11.0
1968 1.3 14 2.6 12.5
4 (63) 1968 13.2 100 – 10.5
1969 4.7 3 7.2 13.7

from 15% to 2.5%, and an increase in serum comple- FLUDARABINE


ment levels toward normal. Cessation of chlorambu-
cil was followed by relapse, and a second course Jacobs388 described a patient with idiopathic CAS
again produced marked improvement. The patient who initially responded to cyclophosphamide-con-
subsequently was able to maintain an adequate taining chemotherapy but subsequently became
hematocrit level without therapy. refractory to this agent. Treatment was changed to
Ramos and associates386 described a 54-year-old fludarabine which was given intravenously daily for
female with a high thermal amplitude, low-affinity 5 days and repeated every 28 days for a total of three
IgG anti-Pra cold autoantibody who did not respond courses. This resulted in a clinical remission with a
to prednisone, splenectomy, IVIG, high-dose intra- normal hemoglobin, reticulocyte count, and bilirubin
venous pulse corticosteroids, and cyclophosphamide. that had lasted 4 years by the time of publication.
She required daily transfusions. She finally responded This is the first report of the use of fludarabine in
21 days after receiving daily plasma exchange (×3), idiopathic CAS, although the drug has been used for
with pulse cyclophosphamide on the third day, fol- patients with WAIHA associated with chronic lym-
lowed by escalating daily oral cyclophosphamide. phocyte leukemia.389 Therapy was successful in only
Chaplin387 reported on two patients with primary one of eight patients, and five other patients devel-
chronic CAS who were treated with chlorambucil for oped AIHA after fludarabine therapy despite a
10 and 7.5 years with cumulative doses of 14 g and continuing response of their leukemia. Jacobs388
6.6 g, respectively. Both responded favorably; however, nevertheless suggested that consideration should be
malignant lymphoma eventually developed in both given to the use of fludarabine for patients with
patients. The authors attempted to assess the incidence idiopathic CAS whose disease is refractory to con-
of lymphoma as part of the natural course of untreated ventional alkylating agents.
CAS but failed for want of proof. They mailed a ques-
tionnaire to 135 United States hematology-oncology
programs and received 77 responses. No lymphomas RITUXIMAB
were reported among patients with chronic CAS, but
one report indicated that a lymphoma developed in Berentsen and colleagues390 reported the use of rituxi-
3 of 141 patients receiving prolonged chlorambucil mab for six patients with CAS. One patient achieved a
therapy for polycythemia vera. complete response defined as disappearance of clini-
In patients who do not respond satisfactorily to cal symptoms, hemolytic anemia, monoclonal IgM
therapy with immunosuppressive drugs, bone marrow kappa band, and morphological and flow-cytometric
depression and temporary worsening of anemia result. signs of clonal lymphoproliferation in the bone
Therefore, it is wise to begin with daily doses of 2 mg marrow. Four partial responses were observed,
or 4 mg per day. Biweekly blood counts (including a including a response to retreatment in one patient.
reticulocyte count) should be obtained, and the daily Two patients were categorized as nonresponders.
dose may be increased by 2 mg every 2 months until Hemoglobin levels increased by a median of 4.1 g/dL
favorable effects or limiting adverse reactions result. in the total group and by 4.7 g/dL in the responders,
The latter effects usually consist of marrow suppres- who also experienced substantial improvements in
sion, although anorexia and nausea could limit dosage clinical symptoms. The treatment was well tolerated.
as well. If a favorable response does result, mainte- The authors concluded that the results were very
nance therapy may be advisable, although an alterna- encouraging.
tive approach is to use therapy intermittently as Pulik and coworkers391 reported on a 77-year-old
needed.10,383 female who had idiopathic CAS with a cold agglutinin
438 Immune Hemolytic Anemias

titer above 2000, which initially was responsive to pred- Gharib and Poynton395 reported on a 53-year-old
nisolone and cyclophosphamide. When a relapse man who had idiopathic CAS with an anti-I cold
occurred, however, treatment with the same drugs did agglutinin titer of 524,288 at 4°C. He was treated with
not improve the hemolysis, and she required multiple cyclophosphamide, azathioprine, prednisolone, and
transfusions. She then received four weekly doses of cyclosporin A without significant symptomatic
rituximab, cyclophosphamide was stopped, and corti- benefit. He then received four doses of rituximab
costeroids were tapered. Red cell transfusions were (375 mg/m2 weekly), after which the cold agglutin in
stopped after the first rituximab infusion, and the titer fell gradually; it became undetectable 9 months
hemoglobin level improved. Hemolysis recurred, later. The patient remains symptom-free 3.5 years
however, and it was then that the authors decided to later, with a normal blood count and reticulocyte
resume treatment with rituximab using a different count.
schedule: one injection every 2 weeks for 2 months, Zaja and associates396 reported that rituximab
then progressively tapered to every 3, 6, and finally, induced a marked B-lymphocyte depletion with
6 weeks. A complete remission was achieved in substantial reduction in the production of cold
6 months and has been maintained with one mainte- agglutinin titer (1:512 to 1:8) in a 72-year-old male
nance injection every 2 months. with CAS. The patient, who was unresponsive to
Lee and associates202 described six patients with conventional immunosuppressive therapies, achieved
AIHA who were treated with rituximab, three of a complete resolution of his hemolytic anemia, with
whom had CAS. One of the patients had previously hemoglobin improving from 7.3 g/dL to 14.3 g/dL.
been described in detail as a 75-year-old female who Morselli and colleagues214 described a patient with
had a non–Hodgkin’s lymphoma of B-cell origin asso- idiopathic mixed warm and cold AIHA who responded
ciated with a monoclonal IgM and CAS.392 Hemolysis to corticosteroids initially but relapsed after the dose of
was not responsive to treatment with corticosteroids, prednisone was reduced to 25 mg/day. Rituximab was
cyclophosphamide, and plasma exchange. She started at a dose of 375 mg/m2 once weekly, for a total
required 12 RBC transfusions during a 1-month of four doses. The hemoglobin improved from 7 g/dL
period. After treatment with rituximab, transfusions to 13.5 g/dL just before the third dose, although bio-
were no longer necessary, paraprotein became unde- chemical signs of hemolysis remained substantially
tectable, and hematocrit increased to 37% with a unaltered. One month after the first cycle of therapy, a
normal reticulocyte count. second course of rituximab was administered at the
Bauduer 393 reported favorable results using same dosage schedule. A complete remission ensued,
rituximab for a patient with CAS associated with a which had been sustained for 7 months at the time of
CD20-positive, low-grade non–Hodgkin’s lym- the report. The patient experienced no side effects of
phoma. The hemoglobin was 8.5 g/dL; the DAT was therapy.
strongly positive with anticomplement serum and Two patients with severe CAS who responded to
weakly positive with anti-IgG; and the cold agglu- rituximab were reported by Engelhardt and co-
tinin titer was 4096. Treatment with oral and workers.397 The first patient, a 50-year-old female,
intravenous cyclophosphamide, prednisolone, cyclo- had been treated with prednisone, cyclophos-
sporine, and plasma exchanges were ineffective, and phamide, mycophenolate mofetil, and plasmaphere-
the patient required transfusion of 33 units of sis before four doses of rituximab were given at
RBCs. After four weekly injections of rituximab at weekly intervals. An increase in the hemoglobin
375 mg/m2, the patient no longer required transfu- level and decreases in bilirubin and LDH levels and
sion, signs of hemolysis abated, and the hemoglobin reticulocytes were noted with the first infusion.
level increased to 12.5 g/dL. Corticosteroids were Mycophenolate mofetil was discontinued, and the
tapered gradually and then discontinued. The remis- patient had maintained her remission for 9 months at
sion had been maintained for 9 months at the time of the time of the report. The second patient, a 60-year-
publication. old female with idiopathic CAS, had worsening
Layios and colleagues394 reported on a 67-year-old anemia (hemoglobin 10 g/dL) and Raynaud’s
patient who had acrocyanosis, CAS with a hemoglobin phenomenon, for which she was treated with four
of 7.6 g/dL, and an indolent non–Hodgkin’s lym- doses of rituximab. The hemoglobin increased, and
phoma. She was treated with chlorambucil, cortico- 11 months after the last rituximab infusion was
steroids, plasmapheresis, and vincristine without 12.9 g/dL.
significant improvement and required transfusion of For more information regarding potential adverse
2 units of RBC every 3 weeks. She was treated with four effects of rituximab therapy, see the earlier section of
weekly doses of rituximab, and therapy with inter- this chapter on WAIHA.
feron-α2a was begun 2 weeks later. Within 2 months,
she was free of signs of hemolysis, and the remission ALPHA-INTERFERON 2B
lasted for 7 months. Subsequently, however, signs of
hemolysis recurred and continued unabated despite Interferon has been used for patients with CAS with
corticosteroids and danazol until her death, caused by the rationale that the idiopathic form of the disease
a stroke. represents one end of a spectrum of low-grade
Management of Autoimmune Hemolytic Anemias 439

lymphoproliferative disorders.398 O’Connor and col- Rosenfield and Jagathambal301 comment that the
leagues399 described a patient with severe CAS who level of cold agglutinins can be reduced by double-unit
responded within 1 month of initiation of therapy plasmapheresis thrice weekly, but clinical improve-
with α-interferon 2b. Fest and coworkers400 described ment in patients with kappa chain IgM CAS has not
a patient with idiopathic CAS who had continued been very great. Buskard404 states that treatment by
hemolysis in spite treatment with corticosteroids, two plasma exchange of CAS that is resistant to chemother-
bolus injections of cyclophosphamide, intravenous apy has been disappointing; the response is brief, and
immunoglobulin, and plasma exchange. Therapy the reduction in hemolysis is minimal. Neither
with α-interferon 2b resulted in a partial response, Rosenfield and Jagathambal nor Buskard supply any
which was sustained for 2 months after discharge further clinical details.
from the hospital. Rodenhuis and associates405 treated an 85-year-old
female who had chronic CAS associated with an IgM
anti-I with eight weekly plasma exchanges. There
were significant reductions in cold agglutinin titer, but
Plasma Exchange the patient’s clinical condition did not improve, and
There is some logic to the use of plasma exchange in she continued to require blood transfusions.
hemolytic anemias caused by IgM antibodies, as IgM In 1993, McLeod and colleagues298 reviewed 14
has a predominantly intravascular distribution, and reported cases of CAS that had been treated with
experience has been favorable with treatment of IgM- plasma exchange.306-308,405,406 They stated that most
induced hyperviscosity syndrome and hemostatic patients responded with rapid and substantial reduc-
defects in Waldenström’s macroglobulinemia.401,402 tions in antibody titer and that elevations of hemoglo-
The results of plasma exchange for patients with CAS, bin level and/or decreases in transfusion requirement
however, have been far from impressive, and improve- were seen in many patients. The notable exception
ment, when it occurs, has generally been only tempo- was the patient reported by Rodenhuis and associ-
rary.6 Further, dealing with cold agglutinins with a ates.405 Improvement in many cases was transient,
high thermal range necessitates special procedures to however, and apheresis therapy did not confer
prevent autoagglutination during the procedure. significant long-term benefit. In summary, then, the
Isbister and associates401 described results for procedure appears to offer a modest degree of tempo-
three patients with CAS. For the first patient, a 4-liter rary benefit to some patients with CAS.
plasma exchange and institution of corticosteroid
therapy resulted in control of “severe autoimmune Splenectomy
hemolytic anemia.” No cold agglutinin titers are
mentioned. The second patient, with immunoblastic Splenectomy is generally considered ineffective for
lymphadenopathy, had an IgM agglutinin and patients with CAS. McCurdy and Rath reported failure
hemolysin reactive up to body temperature. Cortico- in one patient.85 Bell and colleagues406a described the
steroid therapy and plasma exchange were associ- outcome in three patients; two died of sepsis and one
ated with a fall in agglutinin titer at 4°C from 128 to died of lymphoma. Pirofsky12 described one patient
8, disappearance of autoagglutination at 37°C, and who derived no clinical benefit.
sustained response to blood transfusion. A third Dacie,6,16 however, also described three patients
patient, with a wide thermal range IgM autoagglu- with “atypical” CAS who demonstrated marked
tinin, failed to show any clinical response to plasma clinical improvement after splenectomy. One patient
exchange “despite a fall in antibody titer.” The had a complete and lasting remission (except for a
authors comment that the value of the plasma persistently positive DAT). A second patient had a
exchange procedures in the first two cases is difficult remission that lasted for 2 years, and when hemoly-
to assess. This is particularly true in view of the fact sis recurred, she responded well to corticosteroids.
that no serologic data are given in the first patient, The third patient had a chronic hemolytic anemia
and the second patient, with a cold agglutinin titer of that was influenced favorably by ACTH and sple-
only 128, could have had an atypical WAIHA with a nectomy. Remarkably, these patients had similar
warm hemolysin (likely to respond to corticosteroid unusual serologic features in common. The serum
therapy) rather than CAS. antibodies of all patients failed to cause agglutina-
Logue and colleagues403 reported the results of tion of normal RBCs above 30°C but nevertheless
large-volume plasma exchange performed on two caused lysis of enzyme-treated RBCs at 37°C.
occasions for a patient with CAS. The procedure was Furthermore, one of the sera caused lysis to a higher
performed in a room heated to 37°C. After exchange titer at 37°C than at 20°C (the other two sera were not
of 3000 mL of plasma, there was a 70% fall in cold tested at 20°C). The finding of lysis at 37°C but not
agglutinin level, as measured by the ability of the cold at 20°C suggested that the lysis was being produced
agglutinin and fresh serum to lyse RBCs of a patient by a factor that was distinct from the cold agglu-
with paroxysmal nocturnal hemoglobinuria. Within tinin. That warm hemolysins (i.e., optimally active
1 week, however, the antibody level returned to its at 37°C) are unusual in CAS is further emphasized
original parameters. by the study of von dem Borne and coworkers.407
440 Immune Hemolytic Anemias

These authors described warm hemolysins in 69 Danazol


patients; none of the patients had CAS. Lysins reac-
tive at 37°C against trypsinized cells but reactive to a Lugassy and colleagues237 reported on a patient with
higher titer at 20°C do occur in a minority of patients severe primary CAS who also had extensive cuta-
with CAS, however. neous sclerodermic changes. The patient had a mon-
Evans and colleagues408 described a further patient oclonal IgM cold agglutinin with anti-i specificity in
with CAS who had marked improvement lasting for the serum with a titer of 1:1024. The hemoglobin
18 months after splenectomy. This patient’s serum decreased to 5.7g/dL with signs of severe hemolysis.
was also capable of causing lysis at 37°C, whereas The patient was treated with prednisone, 60 mg per
agglutination was negative above 31°C.409 Lysis was day for 1 month, without response. Danazol treat-
more pronounced at 20°C than at 37°C, however, in ment was begun at a dose of 600 mg per day, and
contrast to Dacie’s patients. The serologic findings in within 6 weeks, the hemoglobin rose to 11.0 g/dL and
the patients described by Dacie and Evans are sum- signs of hemolysis diminished. After 4 months of
marized in Table 11-17.16,408,409 danazol therapy, the hemoglobin level was 12.5 g/dL.
Because the total number of patients with CAS No changes were observed in the cutaneous sclero-
who have had a favorable response to splenectomy derma. Additional favorable effects of danazol were
is small, it is impossible to draw firm conclusions described in four patients with CAS.410
regarding the significance of the unusual serologic
features observed. Nevertheless, it is evident that
splenectomy has benefited only a small number of
patients with CAS, and all these patients had
Therapeutic Failures
hemolysins reactive at 37°C. It is conceivable that Although chronic idiopathic CAS is often a mild disor-
the serologic features described in these patients der, there are multiple reports of more serious disease
could be of some value in selecting patients for the and the failure of a variety of therapeutic maneuvers.
operation. Ramos and coworkers411 reported on a patient with an
IgM bithermic anti-ITP who failed to respond to oral
steroids, oral cyclophosphamide, high-dose pulse
Intravenous Immunoglobulin intravenous corticosteroids, and plasma exchanges. He
died of sepsis following an unsuccessful trial of chlo-
Relatively few data are available concerning the use of rambucil. Autopsy revealed an unsuspected dissemi-
IVIG in CAS. A review of cases of AIHA treated with nated non–Hodgkin’s lymphoma.
IVIG was developed by the University HealthSystem Seldon and associates412 reported a fatal case of
Consortium in 1999.258 Of the 77 cases of AIHA acute low-titer wide-thermal-range CAS. High-dose
evaluated, all but one were WAIHA. The exception corticosteroids, cyclophosphamide, and plasmaphere-
was a patient with CAS who was refractory to this sis failed to control hemolysis. Similarly, Rousey and
therapy.260 Smith413 reported on a patient who had idiopathic

TABLE 11-17. SEROLOGIC FINDINGS IN FOUR PATIENTS WITH COLD AGGLUTININ SYNDROME WHO
RESPONDED TO SPLENECTOMY

Lysis
Agglutination Normal Cells Normal Cells (pH 6.5–7.0) Enzyme-Treated Cells

Age, 2°C–4°C 20°C 31°C 20°C 37°C 20°C 37°C


Reference Sex (Titer) (Titer) (Titer) (Titer) (Titer)

Dacie16 73 512 32 0 0 0 NT* 32


F
Dacie16 56 1,024 32 0 0 0 NT* 32
F
Dacie16 32 256 32 0 0 0 0 64
F
Evans et al.408,409 57 500,000 NT* 0 2+ 1+ NT* NT*
M

* Not tested.
Management of Autoimmune Hemolytic Anemias 441

CAS with a low-titer IgM cold agglultinin with anti-Pr Plasmapheresis has been reported to be successful
specificity, and who died because of refractory hemo- in a case described by Roy-Burman and Glader.418 A
lysis. The patient required frequent transfusions (2 5-year-old girl was found to have a biphasic
to 5 units of RBC weekly). All therapies that were hemolysin and a hematocrit of 22%. She required nine
attempted failed, including prednisone, cyclophos- transfusions over the course of 7 days, with her hema-
phamide, azathioprine, IVIG, plasmapheresis, and tocrit reaching a nadir of 11% on the fifth hospital day.
danazol. Serologic studies documented rising titers of She also received corticosteroids and IVIG. A 3-day
the IgM cold agglutinin, and immediately prior to course of plasmapheresis was administered, and once
death, the thermal amplitude reached 37°C. it was initiated, she required no further transfusion.
Hillen and Bakker414 reported failure of interferon- On the basis of this one case, the authors speculate
α-2b therapy in three patients with CAS that was not that early clearance of the offending antibody might
associated with concomitant disease, except that one be possible with plasmapheresis in PCH, as the
patient showed a diffuse infiltration with a small disease is transient and significant rebound of anti-
number of monoclonal lymphoplasmacytic cells, com- body might not occur.
patible with an early phase of macroglobulinemia or We are aware of the results of splenectomy in only
lymphoma. This clonal proliferation did not change in two patients. Both patients had chronic PCH, and,
subsequent biopsies during 3 years of follow-up. surprisingly, both benefited from the procedure.
Treatment was not very well tolerated, and patients Banov417 described a 51-year-old male who had fre-
complained of anorexia, weight loss, fatigue, fever, quent episodes of hemoglobinuria after cold exposure
myalgia, and headache. Therapy had to be discontin- over a period of 10 years. He then sustained a splenic
ued for one patient after 7 months of weight loss of laceration as a result of an automobile accident, and
greater than 10%. splenectomy was therefore performed. He had no
Five patients with idiopathic chronic CAS who failed further episodes of hemoglobinuria despite continued
to respond to cladribine were reported by Berentsen exposure to cold. It was not until 4 years later during
and coworkers.415 The drug was administered as a hospitalization for pneumonia that his intriguing
2-hour intravenous infusion of 0.14 mg/kg/day for history led to the performance of laboratory tests that
5 consecutive days. A second course was given after 12 documented the presence of a positive Donath-
weeks. Two patients received one course of therapy, Landsteiner reaction and serologic evidence for
two received two courses, and one patient received six syphilis. The authors speculated that reduction in
courses with a 1-month interval between the last four total antibody formation was a possible explanation
courses. None of the patients achieved a stable for the patient’s improvement.
improvement of clinical symptoms or hemoglobin and The other patient who responded well to splenec-
IgM levels. Although there were no serious side effects tomy was reported by Ries and coworkers418a (see
of therapy, four patients developed some degree of Chapter 3, page 78).
leukopenia and neutropenia, and three patients devel-
oped lymphopenia.
SECONDARY AUTOIMMUNE
PAROXYSMAL COLD HEMOLYTIC ANEMIAS
HEMOGLOBINURIA
Chronic Lymphocytic Leukemia
Acute postinfectious forms of paroxysmal cold hemo- INCIDENCE OF AIHA IN CLL
globinuria (PCH) occur in children and terminate
spontaneously.416 Accordingly, the patients require AIHA is the most frequent autoimmune disorder
only supportive care. During the acute phase of the described in chronic lymphocytic leukemia (CLL),
disease, severe intravascular hemolysis is usual, and and conversely, CLL is the hematologic malignancy
transfusion of RBC may be necessary. Cortio- in which AIHA occurs most frequently (also see
costeroids are often given, although their effectiveness Chapter 3).7
is difficult to evaluate because of the transient nature Mauro and colleagues419 reported on 52 patients
of the hemolysis. with AIHA observed within a series of 1203 patients
Chronic PCH is very rare, and in these patients, pro- (4.3%) with CLL. Nineteen were observed at the time of
tection from cold is important in preventing attacks of CLL diagnosis, and 33 were observed during the clini-
hemolysis. There are many reports in earlier literature cal follow-up. Some 90% of the patients with AIHA
of clinical improvement caused by antisyphilitic drugs showed active CLL, and 25% had been treated previ-
in chronic PCH associated with syphilis, although not ously. A higher lymphocyte count, older age, and male
all patients responded.417 Data concerning the use of gender were linked significantly with an increased rate
corticosteroids in chronic PCH are too scanty to allow of AIHA at CLL diagnosis (Table 11-18).419 An IgG
for conclusions concerning their effectiveness. autoantibody was present in 87% of cases, and IgM was
442
TABLE 11-18. PROGNOSTIC SIGNIFICANCE OF FACTORS AT CLL DIAGNOSIS RELATED TO THE OCCURRENCE OF DAT-NEGATIVE
AND DAT-POSITIVE ANEMIA: UNIVARIATE AND MULTIVARIATE ANALYSIS

DAT-Negative Anemia* DAT-Positive Anemia†

Univariate Multivariate Univariate Multivariate

Factors at Percentage with Neg. P P Odds Ratio Percentage with Pos. P P Odds Ratio
CLL Diagnosis N DAT Anemia Value Value (95% CI) N DAT Anemia Value Value (98% CI)

Age (yr)
≤65 692 7.2 650 1.2
>65 492 9.5 .15 .04 1.58 456 2.4 .14 .01 3.18
(1.02–2.45) (1.19–8.52)
Sex
M 718 7.9 678 2.5
F 486 8.5 .69 .4 — 428 0.4 .01 .01 0.16
(0.03–0.73)
Lymph × 109/L
≤60 1072 5.7 1016 0.5
>60 112 31.0 <.00001 <.00001 17.84 90 14.0 <.00001 <.00001 22.7
(4.84–12.7) (8.23–62.6)

* Patients with DAT-positive anemia (AIHA) at CLL diagnosis excluded (19 patients).
† Patients with DAT-negative anemia at CLL diagnosis excluded (97 patients).
From Mauro FR, Foa R, Cerrett R, Giannarelli D, Coluzzi S, Mandelli F, et al: Autoimmune hemolytic anemia in chronic lymphocytic leukemia: Clinical, therapeutic, and prognostic features. Blood
2000;95:2786–2792.
Management of Autoimmune Hemolytic Anemias 443

present in 13%. In the patients with IgM autoantibod- ures. In four nonresponding patients, an inadequate
ies, titers at 4°C ranged from 1:256 to 1:8192, and all reticulocytosis was recorded and, the corresponding
were reactive up to 37°C. marrow was hypocellular with dysplastic erythro-
poiesis; three of these patients had previously been
treated with chlorambucil and prednisone. In the
THERAPY other four patients, the hematologic picture was dom-
All patients reported by Mauro and coworkers419 were inated by the presence of active CLL.
treated with prednisone (1–2 mg/kg). In addition, Recurring infections represented the main impedi-
previously untreated patients with other clinical signs ment to use of alternative therapies. Splenectomy was
of active CLL as defined by National Cancer Institute not performed because of medical contraindications
criteria (high and progressive lymphocyte count, in the eight unresponsive patients. For three patients
massive increase of nodal and/or spleen size, etc.) only, a second-line therapy was attempted consisting
received chlorambucil at a dose of 10 mg/m2/day for of high-dose IVIG with prednisone plus azathioprine
6 consecutive days monthly.420 Three patients devel- or cyclosporine. All three patients died without
oped AIHA after fludarabine therapy (see Chapter 8). improvement in AIHA. The eight patients who were
therapeutic failures died after a median time of
2 months, mainly due to infection.
RESPONSE
Fifty of the 52 patients were assessed for response to COMPLICATIONS AND OUTCOME
therapy; two were lost to follow-up. Four patients with
stable CLL were treated with prednisone alone, and the Eight of the 35 patients who had obtained a CR of
remaining 46 with active CLL were treated with pred- AIHA relapsed after a median time of 19 months from
nisone and chlorambucil. Complete response (CR) was the achievement of the response (range, 6–45 months).
defined as patients with no detectable autoantibody A new response of AIHA was achieved by all seven
and persistent hemoglobin values of 12 g/dL or higher. evaluable patients (three obtaining CR).
Partial response (PR) was defined as persistence of the The occurrence of infections represented the main
autoantibody but with a hemoglobin increase to cause of morbidity and mortality. Twenty-seven
12 g/dL or higher or of at least 3 g/dL. Patients with patients (52%) developed infections, most commonly
persistent autoantibody in the absence of a significant a pneumonia. Nine patients experienced steroid-
hemoglobin increase (<3 g/dL) were considered as related hyperglycemia requiring oral hypoglycemic
“failures.” agents or insulin.
Forty-two patients (84%) achieved a response (CR At multivariate analysis, two independent factors
or PR). The median time to reach hemoglobin values appeared significantly related to survival probability of
of 12 g/dL or higher was 4.5 months. Thirty-five CLL patients with AIHA: the Ig class of the autoanti-
patients (70%) obtained a CR, and the actuarial body and the time of AIHA occurrence. A significantly
median time probability to obtain the CR was lower rate of survival was found among patients with
6 months. Eight patients (16%) were considered fail- IgM autoantibody (Fig. 11-10) and those with late

1.0

0.9

0.8

0.7
FIGURE 11-10. Actuarial survival probability of CLL patients
Probability

from AIHA diagnosis according to the Ig class of the auto- 0.6


antibody. Percent surviving at 2 years: IgG (45-patients) 78% lgG
0.5
(SE = 6.6) vs. IgM (7 patients) 21% (SE = 17) (P < .001).
From Mauro FR, Foa R, Cerretti R, Giannarelli D, Coluzzi S, 0.4
Mandelli F, et al: Autoimmune hemolytic anemia in chronic lgM
lymphocytic leukemia: clinical, therapeutic, and prognostic 0.3
features. Blood 2000;95:2786–2792.
0.2

0.1 p < 0.001

0.0
0 12 24 36 48 60 72 84 96
Months
444 Immune Hemolytic Anemias

occurrence of AIHA.419 Patients with IgM autoantibod- of the patients with Evans’s syndrome, and two of the
ies reactive to 37°C have previously been described as patients with autoimmune pancytopenia died after a
indicating a poor clinical outcome.421 follow-up of 4 to 26 months. The authors concluded
that Campath-1H represents an alternative thera-
ALTERNATIVE THERAPY FOR PATIENTS peutic option for severe, refractory autoimmune
WITH AIHA AND CLL cytopenias.
Gupta and colleagues434 used a combination of
Diehl and Ketchum140 emphasize that in some rituximab, cyclophosphamide, and dexamethasone for
patients, AIHA and CLL should be treated independ- eight CLL patients with AIHA refractory to steroids. All
ently, and that treatment with chlorambucil,422 fludar- eight patients achieved a remission of their AIHA.
abine,389 and 2-chlorodeoxyadenosine423,424 have been Median pretreatment hemoglobin was 8.3 g/dL, and
used successfully to control AIHA in otherwise refrac- post-treatment hemoglobin was 14.3 g/dL. The DAT
tory patients. Splenectomy for well-defined AIHA in became negative in five patients. Median duration of
CLL has only rarely been reported,72,425,426 although response was 13 months, and retreatment was also
favorable results have been reported among patients effective in achieving a response on relapse of AIHA.
with less well characterized cytopenias.425,427-429 The authors concluded that a rituximab-based combi-
Results of treatment with IVIG have been reported in nation regimen is highly effective in treating steroid
18 patients, almost always in association with cortico- refractory AIHA of CLL.
steroid therapy.260,430 Lymph nodes and splenomegaly Zaja and coworkers435 treated seven patients with
were noted to decrease with IVIG,430 but impro- CAS with rituximab and reported a complete hemato-
vement in hemoglobin was almost always transient, logic remission associated with a decrease in the
and maintenance IVIG was needed every 3 to agglutinin titer in one patient.
4 weeks.260 Cyclosporine therapy has been reported to Seipelt and associates206 reported a 65-year-old
produce responses in five patients with AIHA and male with CLL that was refractory to initial treat-
CLL.181,182,184,185 The remissions were durable as long ment with fludarabine and epirubicin and 2-chloro-
as the cyclosporine was continued. Both patients who deoxyadenosine. The patient also developed Evans’s
were tapered to less than 3 mg/kg/day had a relapse. syndrome, which responded partially to cyclophos-
Sigler and colleagues431 reported an excellent phamide, vincristine, and prednisone. Subsequently,
response to treatment with vincristine-loaded plate- lymphocytes increased further and cyclophos-
lets for a patient whose AIHA had not responded to phamide was again given, but without effect on the
prednisone, high-dose methylprednisolone, IVIG, peripheral lymphocyte count. At this stage, therapy
vincristine, cyclophosphamide, and cyclosporine. with rituximab (375 mg/m2 four times weekly) was
Four months after the first remission of AIHA, hemol- initiated. The platelet count normalized within
ysis recurred but again responded promptly to 1 week after the first dose of rituximab, and the
another infusion with vincristine-loaded platelets. lymphocyte count dropped within 2 months.
Campath-1H is a humanized IgG monoclonal anti- Chemnitz and colleagues208 also have reported suc-
body specific for the CD52 antigen present on human cessful treatment of steroid- and cyclophosphamide-
lymphocytes and monocytes. The main effect of resistant hemolysis in CLL with rituximab.
Campath-1H is on T cells, and it results in a prolonged
and profound depletion of the CD4 and CD8 subpop-
ulations, particularly the CD4 population.432 It is a Systemic Lupus Erythematosus
potent therapeutic agent against advanced CLL and
immune cytopenias. Rodon and coworkers433 des- Several case series10,436-443 have shown that AIHA
cribed a 70-year-old man with CLL progressive on occurs in approximately 10% of patients with SLE (see
fludarabine therapy, who also had life-threatening Chapter 3), although McDonagh and Isenberg444 did
anemia related to immune hemolysis and pure red cell not identify any patients with AIHA among 215
aplasia. After treatment with Campath-1H, the patient patients with SLE who were followed up for a period
had a sustained complete remission of both CLL and of 2 to 18 years.
anemia, although he died of recurrent sepsis and Kokori and coworkers436 studied 41 consecutive
cachexia 10 months after completion of the treatment. patients with SLE who had 50 clinically important
Willis and associates432 described 21 patients with episodes of AIHA, including 27 patients (66%) in
severe and life-threatening autoimmune cytopenias whom AIHA was the initial disease manifestation. All
resistant to standard immunosuppression who were patients had WAIHA with IgG autoantibodies. All
treated with the monoclonal antibody. The patients patients were treated with corticosteroids, and for
included four who had WAIHA, three with Evans’s nine patients who had concomitant renal involve-
syndrome, and three with autoimmune pancytopenia. ment, cyclophosphamide was used also. One addi-
Three of the patients with WAIHA had a complete or tional patient was given cyclophosphamide when she
partial remission, and all of the patients with Evans’s failed to respond to corticosteroids. Azathioprine was
syndrome or autoimmune pancytopenia had a tran- used for six patients as second-line therapy. The
sient remission. One of the patients with WAIHA, two overall recurrence rate was only 4 per 100 person-
Management of Autoimmune Hemolytic Anemias 445

years, with an expected recurrence-free proportion of lipin antibodies are found in the sera of patients with
83% at 5 years after the initial episode of hemolysis SLE, primary antiphospholipid antibody syndrome,
(Fig. 11-11).436 and several other diseases.442,443 Patients with AIHA
Because AIHA so commonly occurs as an initial who fulfill the criteria of SLE could represent an
manifestation of SLE, the authors suggested that intermediate group of patients who have some mani-
testing for antinuclear antibodies and other lupus- festations of the anticardiolipin syndrome (e.g.,
related autoantibodies is warranted for patients who thromboses).436 Some investigators have reported that
present with AIHA. 436 Patients with SLE who anticardiolipin antibodies act as anti-RBC autoanti-
present with AIHA, when compared with controls bodies in SLE patients with active hemolysis and have
with other types of anemia at the onset of SLE, had identified these antibodies to be mostly of the IgG
an increased risk of renal involvement, thrombocy- isotype.454
topenia, and (possibly) thrombotic episodes during
follow-up. Hodgkin’s Disease
The older medical literature contains a number of
reports of favorable responses of AIHA to sple- AIHA among patients with Hodgkin’s disease is quite
nectomy without adversely affecting the course of unusual, with a reported association of 1.7–2.7%.455,456
the underlying SLE.445-450 For example, Sarles and Accordingly, there are no large studies of the effec-
Levin445 reported on three patients with AIHA and tiveness of various approaches to therapy.
SLE in whom hematologic improvement did not A detailed description of results of therapy among
occur during treatment with steroids but did ensue nine patients with Hodgkin’s disease and WAIHA
following splenectomy. On the other hand, Rivero and has been reported by Eisner and associates.456 All
colleagues451 found that the operation produced only patients responded well to various forms of manage-
short-term benefit in the management of hemocy- ment, including corticosteroids, splenectomy, and
topenic episodes in SLE and stated that it seems war- splenic irradiation. DATs became negative before
ranted only as an emergency procedure for patients death in all of the patients who died of disseminated
unresponsive to medical treatment. disease, and AIHA was not present in the terminal
It is not certain whether AIHA portends a more stages of the disease.
benign or a more severe prognosis for patients with Levine and colleagues457 reported that splenectomy
SLE. Two reports have indicated that AIHA was asso- or corticosteroids plus vincristine were effective in
ciated with increased mortality in univariate analyses, slowing the hemolytic process for two patients, but
although multivariate analyses did not indicate neither of these therapeutic modalities resulted in
hemolytic anemia as an independent predictor of conversion to a negative DAT; however, definitive
mortality.441,452 Other studies either found no associa- chemotherapy for the underlying Hodgkin’s disease
tion of AIHA with adverse outcomes or suggested in these patients (and in a third who received no other
that it could be associated with a more benign therapy for AIHA) resulted in a disappearance of the
course.440 positive DAT. Numerous other authors also have
Patients with AIHA and SLE were more likely to reported that effective therapy for the treatment of
have IgG anticardiolipin antibodies than patients with Hodgkin’s disease results in resolution of the
SLE who had other causes of anemia.436 Anticardio- AIHA.161,455,458-462

1.0

0.9
Proportion without recurrence

0.8

0.7

FIGURE 11-11. Kaplan-Meier plot for the probability of recurrence of 0.6


autoimmune hemolytic anemia in patients with systemic lupus erythe-
matosus. There were 41 patients at baseline, 25 who remained at 0.5
risk at 2 years, 15 at risk at 4 years, and 12 at risk at 6 years. (From
Kokori SI, Ioannidis JP, Voulgarelis M, Tzioufas AG, Moutsopoulos HM: 0.4
Autoimmune hemolytic anemia in patients with systemic lupus
erythematosus. Am J Med 2000;108:198–204.) 0.3

0.2

0.1

0 2 4 6
Time from first episode of autoimmune hemolytic anemia (years)
446 Immune Hemolytic Anemias

Non–Hodgkin’s Lymphoma and other Carcinoma


Lymphoproliferative Disorders AIHA occurs more frequently among patients with car-
Economopoulos and associates463 found that, among cinoma than can be accounted for by chance alone.471 A
370 patients with non–Hodgkin’s lymphoma, 23 number of studies have indicated that removal of the
(6.2%) had AIHA. There were 19 cases of WAIHA tumor led to remission of the AIHA, which relapsed
(5.1%) and 4 (1.1%) with CAS. They reported results with the appearance of metastases.12,472-478 If therapy
of therapy for the four patients with CAS and con- directed against the underlying tumor is ineffective in
cluded that a combination of alkylating agents with resolving the hemolysis, the AIHA should be treated
corticosteroids or even more intensive chemother- independently according to the principles already
apy (in cases with aggressive clinical behavior) reviewed.
appears to be the treatment of choice.
Cases of PCH also have been reported in association Mycoplasma Pneumoniae Infections and
with non–Hodgkin’s lymphoma; combination chemo- Infectious Mononucleosis
therapy for the lymphoma caused a remission in both
the lymphoma and the AIHA.464,465 In some forms of secondary AIHAs, a spontaneous
There are only a few reports of the association remission can be predicted confidently (e.g., hemolytic
between hairy cell leukemia and WAIHA.466 Main- anemias associated with Mycoplasma pneumoniae infec-
waring and colleagues467 reported two patients with tions and infectious mononucleosis). Cold antibodies
fatal CAS complicating hairy cell leukemia. One with a high thermal maximum are always present with
patient died before therapy could be started, and the the former and often present with the latter, so that
other patient was unresponsive to intravenous keeping the patient warm is a logical and uniformly
methylprednisolone and cyclophsophamide, plasma recommended therapeutic maneuver.481-484 For patients
exchanges, interferon, IVIG, and oral cyclosporine. with severe anemia, the hospital room should be
Cesana and coworkers468 reported a patient with warmed to as high a temperature as is tolerable, and
hairy cell leukemia and AIHA who was treated with mittens and warm slippers should be worn as well. One
prednisone yielding temporary improvement of the group even resorted to the use of a NASA space suit to
hemolysis. Subsequent treatment with interferon-α provide constant warmth for a rare patient with pro-
resulted in resolution of both the AIHA and the found disease.485,485a These measures prevent acute exa-
hairy cell leukemia. The AIHA relapsed during cerbations that result from exposure to cold, although
temporary interferon-α withdrawal. The authors more severely affected patients—whose disease is
commented that although the safety of interferon-α perhaps associated with antibodies of particularly high
in hairy cell leukemia–related immune hemolysis thermal amplitude—might have continued hemolysis
needs further confirmation, the case stresses the and progressively more severe anemia even while being
importance of treating the underlying disease in sec- kept as warm as possible. Therefore, close observation
ondary forms of AIHA, even if the appropriate drug and consideration of additional therapy are warranted.
is contraindicated per se for AIHA. Several reports attest to the effectiveness of corticos-
Cohen and associates469 reported a patient with teroid therapy in CAS associated with M. pneumoniae
refractory and transfusion-dependent CAS secondary infection.486-488 As pointed out by Chu and associates,486
to an indolent B-cell lymphoma. She was treated with however, hemolytic anemia due to cold agglutinins in
rituximab (four weekly injections) and achieved com- patients with M. pneumoniae infection is self-limited, so
plete remission with a significant improvement in that the contribution of the corticosteroid therapy to
hemolysis and also became transfusion independent recovery is difficult to ascertain. Nevertheless, it is
with a follow-up of more than 1 year. difficult to resist the temptation to give corticosteroids,
Mori and colleagues470 reported a case of lym- as complications are minimal in a short-term illness.
phoplasmacytoid lymphoma associated with a Specific antimicrobial agents for M. pneumonie have
monoclonal IgM, who also had CAS and thrombo- been recommended.484 Indeed, antibiotics have pro-
cytopenia. He became refractory to prednisolone duced more impressive results than corticosteroids for
and combination chemotherapy and was therefore those patients who still have evidence of unresolved
treated with rituximab. The CAS ameliorated, and pneumonia at the time of development of hemolytic
the serum level of IgM decreased in association with anemia,489 but in many cases, the infection will have
the disappearance of lymphoma cells and the clonal resolved before the onset of the hemolytic anemia.481
rearrangement of the Ig heavy chains in the bone Corticosteroids have been reported to be of distinct
marrow. value in the management of patients with hemolytic
Management of Autoimmune Hemolytic Anemias 447

anemia associated with infectious mononucleo- very significant adverse effects of long-term corti-
sis.486,490-493 Tonkin and colleagues493 reported three costeroid therapy, even at moderate doses. A
patients with severe acute hemolytic anemia. In one common therapeutic error is the prolonged admi-
patient, steroids were not used, and the hemolysis nistration of prednisone (i.e., longer than 4 to
resolved spontaneoulsy over the course of 7 weeks. The 6 months) at a dose of over 15 mg per day to main-
two other patients were treated with corticosteroids and tain a partial remission.
demonstrated marked improvement within a week. 6. Intravenous immunoglobulin and plasma
Keyloun and Grace492 reported a patient who was exchange have modest benefits that are generally
started on therapy with corticosteroids after 2 weeks of of short duration. Data regarding the effectiveness
illness; prompt improvement was noted, with deferves- of danazol is sparse in the recent medical litera-
cence of the patient’s fever, relief of airway obstruction ture. The adverse effects of these therapies are
caused by lymphoid enlargement, a general feeling of minimal, and consequently they are frequently
well-being, and improvement in the hematocrit level. A used as temporizing measures.
less dramatic effect of corticosterold therapy was noted 7. Splenectomy should be considered if the response
by Bowman and coworkers.490 In their patient, therapy to corticosteroids is inadequate, because splenec-
with 60 mg of prednisone resulted in the stabilization of tomy carries the potential for complete and long-
the hematocrit level at 22%, but there was no improve- term remission, and available data suggest that it
ment for the subsequent 9 days. In spite of the reported achieves this aim in more than 50% of patients.
effectiveness of corticosteroids, many patients do not The decision to perform a splenectomy should not
require them, as early spontaneous recovery can be be delayed until adverse effects of corticosteroids
expected with confidence.482,494 become evident but instead should be made early
Geurs and associates308 reported success of plasma- enough to prevent the emergence of such prob-
pheresis in a patient with corticosteroid-resistant lems. Ordinarily, this means making a decision
hemolysis in infectious mononucleosis. The patient within weeks or a few months of diagnosis for
was febrile for 3 weeks and had continuing signs of cases not responding to other therapies.
hemolysis in spite of corticosteroid and antibiotic 8. The major adverse effect of splenectomy is OPSI.
therapies. Finally, plasmapheresis was started, and Education, immunoprophylaxis, and chemopro-
there was a marked and sustained improvement of phylaxis for OPSI must be a perpetual part of
her laboratory values. management of patients in whom splenectomy
has been performed.
9. Immunosuppressive and cytotoxic drugs have
A SUMMARY OF THERAPEUTIC had modest benefits and have significant adverse
PRINCIPLES IN THE MANAGEMENT effects but could be necessary for patients refrac-
OF PATIENTS WITH AUTOIMMUNE tory to other forms of therapy.
10. A rapidly increasing number of reports suggest a
HEMOLYTIC ANEMIA role for newer immunosuppressive drugs, espe-
cially cyclosporine and rituximab.
Idiopathic Warm Antibody Autoimmune 11. Hematopoietic stem cell transplantation is being
Hemolytic Anemia attempted for some autoimmune diseases but is
presently experimental.
1. Initial therapy should be prednisone at a dose of 12. Blood transfusion should be performed when
1.0–1.5 mg/kg/day (or an equivalent dose of necessary, even though the compatibility test
another corticosteroid drug). might reveal gross incompatibility of all units.
2. Higher doses of corticosteroids may be tried, but Special compatibility test procedures are neces-
the potential benefits vs. the potential adverse sary prior to selection of the most appropriate unit
effects of high-dose corticosteroid therapy are not for transfusion (see Chapter 10).
well defined.
3. Responses are often rapid, and it is unusual for
improvement to become evident after 3 weeks of Idiopathic Cold Agglutinin Syndrome
therapy.
4. For patients who respond to treatment, tapering 1. Exposure to cold should be avoided; this may be
of steroid dosage should begin after the hematocit the only therapy necessary even though a chronic
level reaches 30% and should be carried out hemolytic anemia might persist.
slowly over a period of months. 2. The patient should be encouraged to tolerate the
5. If more than 10 mg of prednisone per day (or, at presence of mild or moderate symptoms of anemia
most, 15 mg prednisone per day) is required to because of the high risk-to-benefit ratio of therapeu-
maintain an adequate remission (hematocrit level tic maneuvers that have been used for this disorder.
>30%), therapy with corticosteroids should be con- 3. Chlorambucil (or cyclophosphamide) produces
sidered to have failed. Alternative approaches to benefit for a minority of patients and should be
management should be initiated because of the tried if significant symptoms of anemia are present.
448 Immune Hemolytic Anemias

4. Patients who have anemia with intolerable symp- of infection are still present. The effectiveness of
toms and who do not respond to chlorambucil are corticosteroids is uncertain.
probably best managed by a chronic program of 5. Hemolytic anemia associated with infectious
transfusion. mononucleosis should be treated with cortico-
5. Corticosteroids are generally ineffective for this steroids unless the anemia is mild. If cold reactive
disorder. In a few patients, high doses might cause antibodies with high thermal maximum are
some benefit, but the hazards of prolonged corti- demonstrated, the patient should be kept warm.
costeroid therapy usually greatly outweigh the
benefits. The degree of improvement must justify
the considerable long-term risks. R E F E R E N C E S
6. Plasma exchange offers a modest degree of tempo-
rary benefit to some patients. The procedure is com- 1. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias,
plicated by autoagglutination at room temperature. 1st ed. New York: Churchill Livingstone, 1980.
2. Petz LD: Treatment of autoimmune hemolytic anemias. Curr
7. Splenectomy is generally ineffective, although a Opin Hematol 2001;8:411–416.
few reports of success have been published, parti- 3. Domen RE: An overview of immune hemolytic anemias.
cularly for patients with atypical serologic findings Cleve Clin J Med 1998;65:89–99.
that include the presence of warm hemolysins. 4. Jandl JH: Textbook of Hematology, 2nd ed. Boston: Little,
Brown, 1996
8. Anecdotal reports suggest that rituximab, fludara- 5. Boumpas DT, Chrousos GP, Wilder RL, Cupps TR, Balow JE:
bine, and interferon could be of benefit for some Glucocorticoid therapy for immune-mediated diseases: Basic
patients. and clincal correlates. Ann Intern Med 1993;119:1198–1208.
9. Other therapies are of unproven value. 6. Dacie JV: Auto-immune haemolytic anaemias (AIHA):
Treatment. In The Haemolytic Anaemias, 3rd ed. vol. 3. The
Auto-Immune Haemolytic Anaemias. New York: Churchill
Paroxysmal Cold Hemoglobinuria Livingstone, 1992:452–520.
7. Engelfriet CP, Overbeeke MA, van dem Borne AE: Auto-
1. Most patients with this disorder have an acute and immune hemolytic anemia. Semin Hematol 1992;29:3–12.
transient hemolytic anemia, so that often only sup- 8. Rosse WF: Clinical Immunohematology: Basic Concepts and
Clinical Applications. Boston: Blackwell Scientific, 1990.
portive care is necessary. Frequent monitoring of 9. Gibson J: Autoimmune hemolytic anemia: Current concepts.
the patient’s hematologic status is necessary Aust N Z J Med 1988;18:625–637.
because the hemoglobin and hematocrit can drop 10. Worlledge S: Immune Haemolytic Anaemias. In Hardisty RM,
precipitously. Meticulous attention should be given Weatherall DJ (eds): Blood and Its Disorders. Oxford:
to keeping the patient warm. Blackwell Scientific, 1982:479–513.
11. Zupanska B, Sylwestrowicz T, Pawelski S: The results of
2. The severity of hemolysis could dictate the need for prolonged treatment of autoimmune haemolytic anaemia.
blood transfusion (see Chapter 10). Haematologia 1981;14:425–433.
3. A short course of corticosteroid therapy is war- 12. Pirofsky B: Autoimmunization and the Autoimmune Hemo-
ranted empirically if hemolysis is severe. lytic Anemias. Baltimore: Williams & Wilkins, 1969.
13. Allgood JW, Chaplin H Jr: Idiopathic acquired autoimmune
4. If signs of an infection are present, appropriate hemolytic anemia: A review of forty-seven cases treated from
antibiotics should be provided. 1955 through 1965. Am J Med 1967;43:254–273.
5. In the rare patient with chronic paroxysmal cold 14. Eyster ME, Jenkins DE Jr: Erythrocyte coating substances in
hemoglobinuria, very limited data suggest that patients with positive direct antiglobulin reactions: Corre-
splenectomy might be of benefit. lation of gamma-G globulin and complement coating with
underlying diseases, overt hemolysis and response to therapy.
Am J Med 1969;46:360–371.
Secondary Autoimmune 15. Dameshek W, Komninos ZD: The present status of treatment
of autoimmune hemolytic anemia with ACTH and cortisone.
Hemolytic Anemias Blood 1956;11:648
16. Dacie JV: The Haemolytic Anaemias. Part II. Auto-Immune
1. In patients with AIHA secondary to chronic lym- Haemolytic Anaemias, 2nd ed. London: J&A Churchill, 1962.
phocytic leukemia, the hemolysis often responds 17. Horster JA: Die Korticosteroid-Behandlung Hematologische
well to corticosteroids. Alternative forms of therapy und Verwandter Erkrankungen. Stuttgart: Georg Theime
that have been used include cyclosporine, chloram- Verlag, 1961.
18. Ozsoylu S: High dose intravenous methylprednisolone (HIVMP)
bucil, fludarabine, 2-chlorodeoxyadenosine, IVIG, in hematologic disorders. Hematol Rev 1990;4:197–207.
and splenectomy. Infections represent the main 19. Ozsoylu S: Megadose methylprednisolone for Evans syn-
cause of morbidity and mortality related to treat- drome. Pediatr Hematol Oncol 2000;17:725–726.
ment of the immune hemolysis. 20. Meyer O, Stahl D, Beckhove P, Huhn D, Salama A: Pulsed
high-dose dexamethasone in chronic autoimmune haemolytic
2. Hemolysis associated with SLE generally res- anaemia of warm type. Br J Haematol 1997;98:860–862.
ponds well to therapy described for idiopathic 21. Meytes D, Adler M, Viraq I, Feigl D, Levene C: High-dose
WAIHA. methylprednisolone in acute immune cold hemolysis. N Engl
3. The most effective therapy for AIHA in patients with J Med 1985;312:318.
lymphomas is treatment of the underlying disorder. 22. Hari P, Srivastava RN: Pulse corticosteroid therapy with
methylprednisolone or dexamethasone. Indian J Pediatr
4. Cold agglutinin syndrome associated with M. pneu- 1998;65:557–560.
moniae infection should be treated by keeping the 23. Leddy JP, Swisher SN: Acquired Immune Hemolytic
patient warm and administering antibiotics if signs Disorders (Including Drug-Induced Immune Hemolytic
Management of Autoimmune Hemolytic Anemias 449

Anemia). In Samter M (ed): Immunologic Diseases. Boston: 49. Tanoue K, Okita K, Akahoshi T, et al: Laparoscopic
Little, Brown, 1978:1187. splenectomy for hematologic diseases. Surgery 2002;131(1
24. Nordoy A, Neset G: Splenectomy in hematologic diseases. Suppl.):S318–S323.
Acta Med Scand 1968;183:117–126. 50. Antonelli G: Effeti della splenectomia di una particolare forma
25. Lester RS, Knowles SR, Shear NH: The risks of systemic corti- di ittero emolitico acquisto con anemia a tuipo perniciouso.
costeroid use. Dermatol Clin 1998;16:277–288. Polyclinico (med) 1913;13:193–222.
26. Rosse WF: Quantitative immunology of immune hemo- 51. Eppinger H: Die Hepato-Lienallen Erkrankungen. Berlin:
lytic anemia. II. The relationship of cell-bound antibody to Julius Springer, 1920.
hemolysis and the effect of treatment. J Clin Invest 52. Dameshek W, Schwartz SO: Acute hemolytic anemia (acquired
1971;50:734. hemolytic icterus, acute type). Medicine 1940;19:231–327.
27. Evans RS: Autoantibodies in hematologic disorders. Stamf 53. Dameshek W: The management of acute hemolytic anemia
Med Bull 1955;13:152–166. and the hemolytic crisis. Clinics 1943;2:118.
28. Chaplin H Jr: Clinical usefulness of specific antiglobulin 54. Stickney JM, Heck FJ: Primary nonfamilial hemolytic anemia.
reagents in autoimmune hemolytic anemias. Progr Hematol Blood 1948;3:431.
1973;8:25–49. 55. Robson HN: Medical aspects of splenectomy. Edinb Med J
29. Chaplin H, Avioli LV: Grand rounds: Autoimmune hemolytic 1949;56:381.
anemia. Arch Int Med 1977;137:346–351. 56. Welch CS, Dameshek W: Splenectomy in blood dyscrasias.
30. Frank MM, Schreiber AD, Atkinson JP, Jaffe CJ: Pathophy- New Engl J Med 1950;242:601.
siology of immune hemolytic anemia. Ann Int Med 1977; 57. Dreyfus B, Dausset J, Vidal G: Etude clinque et hematologique
87:210–222. de douze cas d’anemie hemolytique acquise avec auto-anti-
31. Kay NE, Douglas SD: Monocyte-erythrocyte interaction in corps. Rev Hemat 1951;6:349.
vitro in immune hemolytic anemias. Blood 1977;50:889–897. 58. Learmonth J: The surgery of the spleen. Br Med J 1951;2:67.
32. Stanbury RM, Graham EM: Systemic corticosteroid therapy— 59. Chertkow G, Dacie JV: Results of splenectomy in auto-
Side effects and their management. Br J Ophthalmol immune haemolytic anaemia. Br J Haematol 1956;2:237.
1998;82:704–708. 60. Young LE, Miller G, Swisher SN: The treatment of hemolytic
33. Bialas MC, Routledge PA: Adverse effects of corticosteroids. disorders. J Chronic Dis 1957;6:307.
Adverse Drug React Toxicol Rev 1998;17:227–235. 61. Crosby WH, Rappaport H: Autoimmune hemolytic anemia. 1.
34. Dequeker J: NSAIDs/corticosteroids—Primum non nocere. Analysis of hematologic observations with particular refer-
Adv Exp Med Biol 1999;455:319–325. ence to their prognostic value; a survey of 57 cases. Blood
35. Caldwell JR, Furst DE: The efficacy and safety of low-dose 1957;12:42.
corticosteroids for rheumatoid arthritis. Semin Arthritis 62. Dausset J, Colombani JL: The serology and the prognosis of 128
Rheum 1991;21:1–11. cases of autoimmune hemolytic anemia. Blood 1959;14:1280.
36. Recommendations for the prevention and treatment of 63. Goldberg A: Radiochromium in the selection of patients with
glucocorticoid-induced osteoporosis: 2001 update. American haemolytic anaemia for splenectomy. Lancet 1966;1:109.
College of Rheumatology Ad Hoc Committee on Gluco- 64. Kinlough RL, Bennett RC, Lander H: The place of splenec-
coritcoid-Induced Osteoporosis. Arthritis Rheum 2001;44: tomy in haematological disorders: The value of 51Cr tech-
1496–1503. niques. Med J Aust 1966;22:1022–1027.
37. Stuck AE, Minder CE, Frey FJ: Risk of infectious complications 65. Mappes G, Fischer J: Experience with splenectomy in hemato-
in patients taking glucocorticosteroids. Rev Infect Dis logic diseases. Dtsch Med Wochenschr 1969;94:584–589.
1989;11:954–963. 66. Schwartz SI, Bernard RP, Adams JT, Bauman AW: Splenec-
38. Piper JM, Ray WA, Daugherty JR, Griffin MR: Corticosteroid tomy for hematologic disorders. Arch Surg 1970;101:338–347.
use and peptic ulcer disease: Role of nonsteroidal anti- 67. Devlin HB, Evans DS, Birkhead JS: Elective splenectomy for
inflammatory drugs. Ann Intern Med 1991;114:735–740. primary hematologic and splenic disease. Surg Gynecol
39. Luo JC, Chang FY, Lin HY, et al: The potential risk factors Obstet 1970;131:273–276.
leading to peptic ulcer formation in autoimmune disease 68. Ahuja S LSSL: Value of surface counting in predicting
patients receiving corticosteroid treatment. Aliment response to splenectomy in haemolytic anaemia. J Clin Pathol
Pharmacol Ther 2002;16:1241–1248. 1972;25:467.
40. Katkhouda N, Mavor E: Laparoscopic splenectomy. Surg Clin 69. Christensen BE: The pattern of erythrocyte sequestration in
North Am 2000;80:1285–1297. immunohaemolysis: Effects of prednisone treatment and
41. Schlinkert RT, Teotia SS: Laparoscopic splenectomy. Arch Surg splenectomy. Scand J Haematol 1973;10:120–129.
1999;134:99–103. 70. Habibi B, Homberg JC, Schaison G, Salmon C: Autoimmune
42. Katkhouda N, Hurwitz MB, Rivera RT, et al: Laparoscopic hemolytic anemia in children: A review of 80 cases. Am J Med
splenectomy: Outcome and efficacy in 103 consecutive patients. 1974;56:61–69.
Ann Surg 1998;228:568–578. 71. Ikkala E, Kivilaakso E, Hastbacka J: Splenectomy in blood
43. Delaitre B, Pitre J: Laparoscopic splenectomy versus open diseases: A report of 80 cases. Ann Clin Res 1974;6:290–299.
splenectomy: A comparative study. Hepatogastroenterology 72. Coon WW: Splenectomy in the treatment of hemolytic
1997;44:45–49. anemia. Arch Surg 1985;120:625–628.
44. Friedman RL, Hiatt JR, Korman JL, Facklis K, Cymerman J, 73. Wilhelm MC, Jones RE, McGehee R, Mitchener JS, Sandusky
Phillips EH: Laparoscopic or open splenectomy for hemato- WR, Hess CE: Splenectomy in hematologic disorders: The
logic disease: Which approach is superior? J Am Coll Surg ever-changing indications. Ann Surg 1988;207:581–589.
1997;185:49–54. 74. Ly B, Albrechtsen D: Therapeutic splenectomy in hematologic
45. Caprotti R, Porta G, Franciosi C, et al: Laparoscopic splenec- disorders: Effects and complications in 221 adult patients.
tomy for hematological disorders: Our experience in adult Acta Med Scand 1981;209:21–29.
and pediatric patients. Int Surg 1998;83:303–307. 75. Akpek G, McAneny D, Weintraub L: Comparative response to
46. Stephens BJ, Justice JL, Sloan DA, Yoder JA: Elective laparo- splenectomy in Coombs-positive autoimmune hemolytic
scopic splenectomy for hematologic disorders. Am Surg anemia with or without associated disease. Am J Hematol
1997;63:700–703. 1999;61:98–102.
47. Rosen M, Brody F, Walsh RM, Tarnoff M, Malm J, Ponsky J: 76. Karpatkin S, Strick N, Siskind GW: Detection of splenic anti-
Outcome of laparoscopic splenectomy based on hematologic platelet antibody synthesis in idiopathic autoimmune
indication. Surg Endosc 2002;16:272–279. thrombocytopenic purpura (ATP). Br J Haematol 1972;23:
48. Katkhouda N, Manhas S, Umbach TW, Kaiser AM: Laparo- 167–176.
scopic splenectomy. J Laparoendosc Adv Surg Tech A 77. McMillan R, Longmire RL, Yelenosky R, Donnell RL,
2001;11:383–390. Armstrong S: Quantitation of platelet-binding IgG produced
450 Immune Hemolytic Anemias

in vitro by spleens from patients with idiopathic thrombocy- 105. Lucas RV, Krivit W: Overwhelming infection in children
topenic purpura. N Engl J Med 1974;291:812–817. following splenectomy. J Peds 1960;57:185.
78. Bowdler AJ: The spleen and hemolytic disorders. Clinics 106. Singer DB: Postsplenectomy Sepsis. In Rosenberg HS,
Haematol 1975;4:231. Bolande RP (eds): Perspectives on Pediatric Pathology.
79. Bowdler AJ: The role of the spleen and splenectomy in autoim- Chicago: Year Book, 1973:285.
mune hemolytic disease. Semin Hematol 1976;13:335–348. 107. Krivit W: Overwhelming postsplenectomy infection. Am J
80. Hughes-Jones NC, Szul L: Determination of the sites of red-cell Hematol 1977;2:193–201.
destruction using 51Cr-labeled cells. Br J Haematol 1957;3:320. 108. Lynch AM, Kapila R: Overwhelming postsplenectomy infec-
81. Jandl JH, Greenberg MS, Yonemoto RH, Castle WB: Clinical tion. Infect Dis Clin North Am 1996;10:693–707.
determination of the sites of red cell sequestration in 109. Brigden ML, Pattullo AL: Prevention and management of
hemolytic anemias. J Clin Invest 1956;35:842. overwhelming postsplenectomy infection—An update. Crit
82. Jandl JH, Jones AR, Castle WB: The destruction of red cells by Care Med 1999;27:836–842.
antibodies in man. I. Observations of the sequestration and 110. Styrt B: Infection associated with asplenia: Risks, mechanisms,
lysis of red cells altered by immune mechanisms. J Clin Invest and prevention. Am J Med 1990;88(5N):33N-42N.
1957;36:1428. 111. Green JB, Shackford SR, Sise MJ, Fridlund P: Late septic
83. Ben-Bassat I, Seligsohn U, Leiba H, Leef F, Chaitchik S, Ramot complications in adults following splenectomy for trauma: A
B: Sequestration studies with chromium-51 labelled red cells prospective analysis in 144 patients. J Trauma 1986;26:999–1004.
as criteria for splenectomy. Israel J Med Sci 1967;3:832. 112. Brigden ML: Overwhelming postsplenectomy infection still a
84. Lewis SM, Szur L, Dacie JV: The pattern of erythrocyte problem. West J Med 1992;157:440–443.
destruction in haemolytic anaemia, as studied with radioac- 113. Holdsworth RJ, Irving AD, Cuschieri A: Postsplenectomy
tive chromium. British Journal of Haematology 1960;6:122. sepsis and its mortality rate: Actual versus perceived risks. Br
85. McCurdy PR, Rath CE: Splenectomy in hemolytic anemia: J Surg 1991;78:1031–1038.
Results predicted by body scanning after injection of Cr51- 114. Schilling RF: Estimating the risk for sepsis after splenectomy in
tagged red cells. New Engl J Med 1958;259:459. hereditary spherocytosis. Ann Intern Med 1995;122:187–188.
86. Najean Y, Cacchione R, Dresch C, Rain JD: Methods of evalu- 115. Schwartz PE, Sterioff S, Mucha P, Melton LJ III, Offord KP:
ating the sequestration site of red cells labelled with 51Cr: A Postsplenectomy sepsis and mortality in adults. JAMA
review of 96 cases. Br J Haematol 1975;29:495–510. 1982;248:2279–2283.
87. Parker AC, MacPherson AIS, Richmond J: Value of 116. Cullingford GL, Watkins DN, Watts AD, Mallon DF: Severe
radiochromium investigation in autoimmune haemolytic late postsplenectomy infection. Br J Surg 1991;78:716–721.
anaemia. Br Med J 1977;1:208. 117. Konradsen HB, Henrichsen J: Pneumococcal infections in
88. Schloesser LL, Korst DR, Clatanoff DV, Schilling RF: splenectomized children are preventable. Acta Paediatr Scand
Radioactivity over the spleen and liver following transfusion 1991;80:423–427.
of chromium 51-labelled erythrocytes in hemolytic anemia. 118. Waghorn DJ, Mayon-White RT: A study of 42 episodes of
J Clin Invest 1957;36:1470. overwhelming post-splenectomy infection: Is current guid-
89. Szur L: Surface counting in the assessment of sites of red cell ance for asplenic individuals being followed? J Infect
destruction. Brit J Haemat 1970;18:591. 1997;35:289–294.
90. Veeger W, Woldring MG, VanRood JJ, et al: The value of the 119. Working Party of the British Committee for Standards in
determination of the site of red cell sequestration in hemolytic Haematology Clinical Haematology Task Force: Guidelines
anemia as a prediction test for splenectomy. Acta Med Scand for the prevention and treatment of infection in patients with
1962;171:507. an absent or dysfunctional spleen. Br Med J 1996;312:430–434.
91. Williams ED, Szur L, Glass HI, Lewis SM, Pettit JE, Ahuja S: 120. Fielding AK: Prophylaxis against late infection following
Measurement of red cell destruction in the spleen. J Lab Clin splenectomy and bone marrow transplant. Blood Rev
Med 1974;84:134–146. 1994;8:179–191.
92. Nightingale D, Prankerd TA, Richards JD, Thompson D: 121. Flegg PJ: Long term management after splenectomy. Br Med J
Splenectomy in anaemia. Q J Med 1972;41:261–267. 1994;308:131.
93. Crosby WH: Splenectomy in hematologic disorders. New 122. Advisory Committee on Immunization Practices: Recommen-
Engl J Med 1972;286:1252. dations of the Advisory Committee on Immunization Practices
94. Dacie JV, Worlledge SM: Auto-immune hemolytic anemias. In (ACIP): Use of vaccines and immune globulins in persons with
Brown EB, Moore CV (eds): Progress in Hematology. New altered immunocompetence. MMWR 1993;42:1–18.
York: Grune and Stratton, 1969:82–120. 123. Deodhar HA, Marshall RJ, Barnes JN: Increased risk of sepsis
95. Pirofsky B: Immune haemolytic disease: The autoimmune after splenectomy. Br Med J 1993;307:1408–1409.
haemolytic anaemias. Clin Haematol 1975;4:167. 124. Hirsh J, Dacie JV: Persistent post-splenectomy thrombocytosis
96. Rosse WF: Quantitative immunology of immune hemolytic and thrombo-embolism: A consequence of continuing
anemia. I. The fixation of C1 by autoimmune antibody and anaemia. Br J Haematol 1966;12:44–53.
heterologous anti-IgG antibody. J Clin Invest 1971;50:727. 125. Visudhiphan S, Ketsa-Ard K, Piankijagum A, Tumliang S:
97. Schwartz SI, Adams JT, Bauman AW: Splenectomy for hema- Blood coagulation and platelet profiles in persistent post-
tologic disorders. Curr Probl Surg 1971;1–57. splenectomy thrombocytosis: The relationship to thromboem-
98. DeWeese MS, Coller FA: Splenectomy for hematologic disor- bolism. Biomed Pharmacother 1985;39:264–271.
ders. West J Surg 1959;67:129. 126. Boxer MA, Braun J, Ellman L: Thromboembolic risk of post-
99. Sandusky WR, Leavell BS, Benjamin BI: Splenectomy: splenectomy thrombocytosis. Arch Surg 1978;113:808–809.
Indications and results in hematologic disorders. Ann Surg 127. Traetow WD, Fabri PJ, Carey LC: Changing indications for
1964;159:695–710. splenectomy. 30 years’ experience. Arch Surg 1980;115:447–451.
100. Sedgwick CE, Hume AH: Elective splenectomy: An analysis 128. Coon WW, Penner J, Clagett P, Eos N: Deep venous thrombo-
of 220 operations. Ann Surg 1960;151:163. sis and postsplenectomy thrombocytosis. Arch Surg
101. Morris DH, Bullock FD: The importance of the spleen in 1978;113:429–431.
resistance to infection. Ann Surg 1919;70:513. 129. Ellison EC, Fabri PJ: Complications of splenectomy. Etiology,
102. Krivit W, Giebink GS, Leonard A: Overwhelming postplenec- prevention, and management. Surg Clin North Am
tomy infection. Surg Clin North Am 1979;59:223–233. 1983;63:1313–1330.
103. King H, Schumacher HB: Splenic studies. I. Susceptibility to 130. Chaffanjon PC, Brichon PY, Ranchoup Y, Gressin R, Sotto JJ:
infection after splenectomy performed in infancy. Am Surg Portal vein thrombosis following splenectomy for hemato-
1952;136:239. logic disease: Prospective study with Doppler color flow
104. Smith CH, Erlandson ME, Schulman I, et al: Hazards of severe imaging. World J Surg 1998;22:1082–1086.
infections in splenectomized infants and children. Am J Med 131. Hayes DM, Spurr CL, Histoff LW, et al: Post splenectomy
1957;22:380. thrombocsytosis. Ann Intern Med 1963;58:259–267.
Management of Autoimmune Hemolytic Anemias 451

132. Bensinger TA, Logue GL, Rundles RW: Hemorrhagic throm- 157. Silva VA, Seder RH, Weintraub LR: Synchronization of plasma
bocythemia; control of postsplenectomy thrombocytosis with exchange and cyclophosphamide in severe and refractory
melphalan. Blood 1970;36:61–69. autoimmune hemolytic anemia. J Clin Apheresis 1994;9:
133. Gluch L, Khouri JM: Pseudohyperkalaemia after splenec- 120–123.
tomy: Pitfall for the unwary. Med J Aust 1994;161:509–510. 158. Ahn YS, Harrington WJ, Byrnes JJ, Pall L, McCrainie J:
134. Parker NE, Jacobs P: Pseudohyperkalaemia—A cause of diag- Treatment of autoimmune hemolytic anemia with Vinca-
nostic confusion. S Afr Med J 1981;60:973–974. loaded platelets. JAMA 1983;249:2189–2194.
135. Ho AM, Woo JC, Kelton JG, Chiu L: Spurious hyperkalaemia 159. Gertz MA, Petitt RM, Pineda AA, Wick MR, Burgstaler EA:
associated with severe thrombocytosis and leukocytosis. Can Vinblastine-loaded platelets for autoimmune hemolytic
J Anaesth 1991;38:613–615. anemia. Ann Intern Med 1981;95:325–326.
136. Erslev AJ: Hypersplenism and hyposplenism. In Beutler E, 160. Medellin PL, Patten E, Weiss GB: Vinblastine for autoimmune
Lichtman MA, Coller BS, Kipps TJ, Seligsohn U (eds): Williams hemolytic anemia. Ann Intern Med 1982;96:123.
Hematology. New York: McGraw-Hill, 2001:683–687. 161. Brady-West DC, Thame J, West W: Autoimmune haemolytic
137. Pearson HA: Red-cell “rubbish” as a key to splenic function. anaemia, immune thrombocytopenia, and leucopenia: An
Lab Mgmt 1982;(September):25–33. unusual presentation of Hodgkin’s disease. West Indian Med
138. Corazza GR, Ginaldi L, Zoli G, Frisoni M, Lalli G, Gasbarrini J 1997;46:95–96.
G, et al: Howell-Jolly body counting as a measure of splenic 162. Vaiopoulos G, Kyriakou D, Papadaki H, Fessas P, Eliopoulos
function: A reassessment. Clin Lab Haematol 1990;12:269–275. GD: Multiple myeloma associated with autoimmune
138a. Holroyde CP, Oski FA, Gardner FH: The “pocked” erythro- hemolytic anemia. Haematologica 1994;79:262–264.
cyte: Red-cell surface alterations in reticuloendothelial imma- 163. Majumdar G, Brown S, Slater NG, Singh AK: Clinical
turity of the neonate. N Engl J Med 1969;281:516–520. spectrum of autoimmune haemolytic anaemia in patients
138b. Reinhart WH, Chien S: Red cell vacuoles: Their size and dis- with chronic lymphocytic leukaemia. Leuk Lymphoma
tribution under normal conditions and after splenectomy. 1993;9:149–151.
Am J Hematol 1988;27:265–271. 164. Dovat S, Roberts RL, Wakim M, Stiehm ER, Feig SA: Immune
139. Zago MA, Covas DT, Figueiredo MS, Bottura C: Red cell pits thrombocytopenia after umbilical cord progenitor cell trans-
appear preferentially in old cells after splenectomy. Acta plant: Response to vincristine. Bone Marrow Transplant
Haematol 1986;76:54–56. 1999;24:321–323.
140. Diehl LF, Ketchum LH: Autoimmune disease and chronic 165. Shastri KA, Logue GL: Autoimmune Hemolytic Anemia. In
lymphocytic leukemia: Autoimmune hemolytic anemia, pure Conn HF (ed): Current Therapy. Philadelphia: WB Saunders,
red cell aplasia, and autoimmune thrombocytopenia. Semin 1997:352–355.
Oncol 1998;25:80–97. 166. Murphy S, LoBuglio AF: Drug therapy of autoimmune
141. Markus H, Forfar JC: Splenic irradiation in treating warm hemolytic anemia. Semin Hematol 1976;13:323–334.
autoimmune haemolytic anaemia. Br Med J (Clin Res Ed) 167. Petz LD: Hemolytic Anemias-Immune. In Conn HF (ed):
1986;293:839–840. Current Therapy. Philadelphia: WB Saunders, 1977:256–260.
142. Fathy N, Furst DE: Combination therapy for autoimmune 168. Rosse WF, Logue GL: Immune hemolytic anemias. Mod Treat
diseases: The rheumatoid arthritis model. Springer Semin 1971;8:379–401.
Immunopathol 2001;23:5–26. 169. Janssen NM, Genta MS: The effects of immunosuppressive
143. Moroni G, Della Casa AO, Ponticelli C: Combination treat- and anti-inflammatory medications on fertility, pregnancy,
ment in autoimmune diseases: Systemic lupus erythematosus. and lactation. Arch Intern Med 2000;160:610–619.
Springer Semin Immunopathol 2001;23:75–89. 170. Leone G, Voso MT, Sica S, Morosetti R, Pagano L: Therapy
144. Ciancio G, Burke GW, Miller J: Current treatment practice related leukemias: Susceptibility, prevention and treatment.
in immunosuppression. Expert Opin Pharmacother 2000;1: Leuk Lymphoma 2001;41:255–276.
1307–1330. 171. Kyle R: Second malignancies associated with chemotherapic
145. Allison AC: Immunosuppressive drugs: The first 50 years and agents. Semin Oncol 1982;9:133.
a glance forward. Immunopharmacology 2000;47:63–83. 172. Park D, Koeffler P: Therapy related myelodysplastic syn-
146. Gonin JM: Maintenance immunosuppression: New agents and dromes. Sem Hematol 1996;33:256–273.
persistent dilemmas. Adv Ren Replace Ther 2000;7:95–116. 173. Feig SA: Second malignant neoplasms after successful treatment
147. Schwartz R, Dameshek W: The treatment of autoimmune of childhood cancers. Blood Cells Mol Dis 2001;27:662–666.
hemolytic anemia with 6–mercaptopurine and thioguanine. 174. Matsuda S, Koyasu S: Mechanisms of action of cyclosporine.
Blood 1962;19:483. Immunopharmacology 2000;47:119–125.
148. Taylor L: Idiopathic autoimmune hemolytic anemia: Response 175. Fathman CG, Myers BD: Cyclosporine therapy for autoim-
of a patient to repeated courses of alkylating agents. Am J mune disease. N Engl J Med 1992;326:1693–1695.
Med 1963;35:130. 176. Emilia G, Messora C, Longo G, Bertesi M: Long-term salvage
149. Hitzig WH, Massimo L: Treatment of autoimmune hemolytic treatment by cyclosporin in refractory autoimmune haemato-
anemia in children with azathioprine (imuran). Blood logical disorders. Br J Haematol 1996;93:341–344.
1966;28:840–850. 177. Kahan BD: Cyclosporine. N Engl J Med 1989;321:1725–1738.
150. Worlledge S: Immune Haemolytic Anemias. In Hardisty RM, 178. Yee GC, Stanley DL, Pessa LJ, et al: Effect of grapefruit juice on
Weatherall DJ (eds): Blood and Its Disorders. Oxford: blood cyclosporin concentration. Lancet 1995;345:955–956.
Blackwell, 1974:714. 179. Kane GC, Lipsky JJ: Drug-grapefruit juice interactions. Mayo
151. Skinner MD, Schwartz RS: Immunosuppressive therapy. 1. Clin Proc 2000;75:933–942.
N Engl J Med 1972;287:221–227. 180. Emilia G, Messora C: Interaction between cyclosporin and
152. Skinner MD, Schwartz RS: Immunosuppressive therapy. 2. chlorambucil. Eur J Haematol 1993;51:179.
N Engl J Med 1972;287:281–286. 181. Dundar S, Ozdemir O, Ozcebe O: Cyclosporin in steroid-
153. Mueller-Eckhardt CH, Kretschmer V: Immunosuppressive resistant auto-immune haemolytic anaemia. Acta Haematol
Therapy in Blood Diseases. New York: Schwabe, 1972:85. 1991;86:200–202.
154. Johnson CA, Abildgaard CF: Treatment of idiopathic autoim- 182. Hershko C, Sonnenblick M, Ashkenazi J: Control of steroid-
mune hemolytic anemia in children: Review and report of two resistant autoimmune haemolytic anaemia by cyclosporine.
fatal cases in infancy. Acta Paediatr Scand 1976;65:375–379. Br J Haematol 1990;76:436–437.
155. Moyo VM, Smith D, Brodsky I, Crilley P, Jones RJ, Brodsky 183. Rackoff WR, Manno CS: Treatment of refractory Evans syn-
RA: High-dose cyclophosphamide for refractory autoimmune drome with alternate-day cyclosporine and prednisone. Am J
hemolytic anemia. Blood 2002;100:704–706. Pediatr Hematol Oncol 1994;16:156–159.
156. Heisel MA, Ortega JA: Factors influencing prognosis in child- 184. Ruess-Borst MA, Waller HD, Muller CA: Successful treatment
hood autoimmune hemolytic anemia. Am J Pediatr Hematol of steroid-resistant hemolysis in chronic lymphocytic leukemia
Oncol 1983;5:147–152. with cyclosporine A. Am J Hematol 1994;46:375–376.
452 Immune Hemolytic Anemias

185. Emilia G, Messora C, Bensi L: The use of cyclosporin-A in the 205. Ahrens N, Kingreen D, Seltsam A, Salama A: Treatment of
treatment of B-chronic lymphocytic leukemia. Leukemia refractory autoimmune haemolytic anaemia with anti-CD20
1995;9:357–359. (rituximab). Br J Haematol 2001;114:244–245.
186. Ferrara F, Copia C, Annunziata M, et al: Complete remission 206. Seipelt G, Bohme A, Koschmieder S, Hoelzer D: Effective
of refractory anemia following a single high dose of treatment with rituximab in a patient with refractory prolym-
cyclophosphamide. Ann Hematol 1999;78:87–88. phocytoid transformed B-chronic lymphocytic leukemia and
187. Friedmann AM, King KE, Shirey RS, Resar LM, Casella JF: Evans syndrome. Ann Hematol 2001;80:170–173.
Fatal autoimmune hemolytic anemia in a child due to warm- 207. Perrotta S, Locatelli F, La Manna A, Cennamo L, De Stefano P,
reactive immunoglobulin M antibody. J Pediatr Hematol Nobili B: Anti-CD20 monoclonal antibody (Rituximab) for
Oncol 1998;20:502–505. life-threatening autoimmune haemolytic anaemia in a patient
188. Duru F, Gurgey A, Cetin M, Kanra T, Altay C: Chronic autoim- with systemic lupus erythematosus. Br J Haematol 2002;116:
mune hemolytic anemia in children: A report of four patients. 465–467.
J Med 1994;25:231–240. 208. Chemnitz J, Draube A, Diehl V, Wolf J: Successful treatment of
189. Allison AC, Eugui EM: Mycophenolate mofetil and its mech- steroid and cyclophosphamide-resistant hemolysis in chronic
anisms of action. Immunopharmacology 2000;47:85–118. lymphocytic leukemia with rituximab. Am J Hematol
190. Gregoor PJHS, van Gelder T, Weimar W: Mycophenolate 2002;69:232–233.
mofetil, Cellcept, a new immunosuppressive drug with 209. McMahon C, Babu L, Hodgson A, Hayat A, Connell NO,
great potential in internal medicine. Neth J Med 2000;57: Smith OP: Childhood refractory autoimmune haemolytic
233–246. anaemia: Is there a role for anti-CD20 therapy (rituximab)? Br
191. Hood KA, Zarembski DG: Mycophenolate mofetil: A unique J Haematol 2002;117:480–483.
immunosuppressive agent. Am J Health Syst Pharm 1997; 210. Seeliger S, Baumann M, Mohr M, Jurgens H, Frosch M,
54:285–294. Vormoor J: Autologous peripheral blood stem cell transplan-
192. Zimmer-Molsberger B, Knauf W, Thiel E: Mycophenolate tation and anti-B-cell directed immunotherapy for refractory
mofetil for severe autoimmune haemolytic anemia. Lancet auto-immune haemolytic anaemia. Eur J Pediatr 2001;160:
1997;350:1003–1004. 492–496.
193. Howard J, Hoffbrand AV, Prentice HG, Mehta A: 211. Schiller G, Tillisch J, Rosen P: Use of anti-CD20 monoclonal
Mycophenolate mofetil for the treatment of refractory auto- antibody for the treatment of Evans syndrome and mono-
immune haemolytic anaemia and auto-immune thrombocy- clonal gammopathy/neuropathy syndrome [abstract]. Blood
topenia purpura. Br J Haematol 2002;117:712–715. 1999;94(Suppl. 1):3536.
194. Maloney DG, Smith B, Rose A: Rituximab: Mechanism of 212. Zompi S, Tulliez M, Conti F, et al: Rituximab (anti-CD20
action and resistance. Semin Oncol 2002;29(1 Suppl. 2):2–9. monoclonal antibody) for the treatment of patients with
195. Grillo-Lopez AJ, Hedrick E, Rashford M, Benyunes M: clonal lymphoproliferative disorders after orthotopic liver
Rituximab: Ongoing and future clinical development. Semin transplantation: A report of three cases. J Hepatol
Oncol 2002;29(1 Suppl. 2):105–112. 2000;32:521–527.
196. Zecca M, De Stefano P, Nobili B, Locatelli F: Anti-CD20 mon- 213. Abdel-Raheem MM, Potti A, Kobrinsky N: Severe Evans’s
oclonal antibody for the treatment of severe, immune- syndrome secondary to interleukin-2 therapy: Treatment with
mediated, pure red cell aplasia and hemolytic anemia. Blood chimeric monoclonal anti-CD20 antibody. Ann Hematol
2001;97:3995–3997. 2001;80:543–545.
197. Coiffier B, Haioun C, Ketterer N, et al: Rituximab (anti-CD20 214. Morselli M, Luppi M, Potenza L, et al: Mixed warm and cold
monoclonal antibody) for the treatment of patients with relaps- autoimmune hemolytic anemia: complete recovery after 2
ing or refractory aggressive lymphoma: a multicenter phase II courses of rituximab treatment. Blood 2002;99:3478–3479.
study. Blood 1998;92:1927–1932. 215. Quartier P, Brethon B, Philippet P, Landman-Parker J, Le Deist
198. Leget GA, Czuczman MS: Use of rituximab, the new FDA- F, Fischer A: Treatment of childhood autoimmune haemolytic
approved antibody. Curr Opin Oncol 1998;10:548–551. anaemia with rituximab. Lancet 2001;358:1511–1513.
199. Byrd JC, Waselenko JK, Maneatis TJ, et al: Rituximab therapy 216. Ng PC, Lee KK, Lo AF, Li CK, Fok TF: Anti B cell targeted
in hematologic malignancy patients with circulating blood immunotherapy for treatment of refractory autoimmune
tumor cells: Association with increased infusion-related side haemolytic anaemia in a young infant. Arch Dis Child
effects and rapid blood tumor clearance. J Clin Oncol 2003;88:337–339.
1999;17:791–795. 217. Zecca M, Nobili B, Ramenghi U, et al: Rituximab for the treat-
200. Ratanatharathorn V, Carson E, Reynolds C, et al: Anti-CD20 ment of refractory autoimmune hemolytic anemia in children.
chimeric monoclonal antibody treatment of refractory Blood 2003;101:3857–3861.
immune-mediated thrombocytopenia in a patient with 218. Hongeng S, Tardtong P, Worapongpaiboon S, Ungkanont A,
chronic graft-versus-host disease. Ann Intern Med 2000;133: Jootar S: Successful treatment of refractory autoimmune
275–279. haemolytic anaemia in a post-unrelated bone marrow trans-
201. Stasi R, Pagano A, Stipa E, Amadori S: Rituximab chimeric plant paediatric patient with rituximab. Bone Marrow
anti-CD20 monoclonal antibody treatment for adults with Transplant 2002;29:871–872.
chronic idiopathic thrombocytopenic purpura. Blood 2001;98: 219. Matthews DC: Ribuximab in the very young: Benefits in
952–957. AIHA, at what risk? Blood 2003;101:3761.
201a. Kakaiya R: New therapy with anti-CD20 antibody (Rituxi- 220. Motto DG, Williams JA, Boxer LA: Rituximab for refractory
mab) for autoimmune hemolytic anemia. Blood Therapies in childhood autoimmune hemolytic anemia. Isr Med Assoc J
Medicine 2003;3:91–96. 2002;4:1006–1008.
202. Lee E, Zamkoff KW, Gentile TC, Zimrin A: Rituxan in the 221. Quartier P, Tournilhac O, Archimbaud C, et al: Enteroviral
treatment of auto-immune hemolytic anemia (AIHA) meningoencephalitis after anti-CD20 (rituximab) treatment.
[abstract]. Blood 2000;96:596–597. Clin Infect Dis 2003;36:e47–e49.
203. Grossi A, Santini V, Longo G, Balestri F, Rossi-Ferrini P, 222. Kunkel L, Wong A, Maneatis T, et al: Optimizing the use of
Morfini M: Treatment with anti-CD20 antibodies of patients rituximab for treatment of B-cell non-Hodgkin’s lymphoma:
with autoimmune thrombocytopenia with or without A benefit-risk update. Semin Oncol 2000;27(Suppl. 12):53–61.
hemolytic anemia; worsening in hemoglobin level [abstract]. 223. Wood AM: Rituximab: An innovative therapy for non-
Blood 2000;96:523. Hodgkin’s lymphoma. Am J Health Syst Pharm 2001;58:215–229.
204. Rai KR, Gupta NK, Janson D, Patel DV, Ahmed I, Kavuru S: 224. Huhn D, von Schilling C, Wilhelm M, et al: Rituximab therapy
Rituximab, cyclophosphamide and decadron combination is of patients with B-cell chronic lymphocytic leukemia. Blood
highly effective in auto-immune hemolytic anemia associated 2001;98:1326–1331.
with chronic lymphocytic leukemia [abstract]. Blood 2000; 225. Reff ME, Carner K, Chambers KS, Chinn PC, Leonard JE,
96:754–755. Raab R, et al: Depletion of B cells in vivo by a chimeric
Management of Autoimmune Hemolytic Anemias 453

mouse human monoclonal antibody to CD20. Blood 1994;83: 249. Dwyer JM: Manipulating the immune system with immune
435–445. globulin. N Engl J Med 1992;326:107–116.
226. Sharma VR, Fleming DR, Slone SP: Pure red cell aplasia due 250. Masson PL: Elimination of infectious antigens and increase of
to parvovirus B19 in a patient treated with rituximab. Blood IgG catabolism as possible modes of action of IVIg. J Auto-
2000;96:1184–1186. immun 1993;6:683–689.
227. Dervite I, Hober D, Morel P: Acute hepatitis B in a patient 251. Yu Z, Lennon VA: Mechanism of intravenous immune globu-
with antibodies to hepatitis B surface antigen who was receiv- lin therapy in antibody-mediated autoimmune diseases. N
ing rituximab. N Engl J Med 2001;344:68–69. Engl J Med 1999;340:227–228.
228. Goldberg SL, Pecora AL, Alter RS, Kroll MS, Rowley SD, 252. Bleeker WK, Teeling JL, Hack CE: Accelerated autoantibody
Waintraub SE, et al: Unusual viral infections (progressive clearance by intravenous immunoglobulin therapy: Studies in
multifocal leukoencephalopathy and cytomegalovirus experimental models to determine the magnitude and time
disease) after high-dose chemotherapy with autologous course of the effect. Blood 2001;98:3136–3142.
blood stem cell rescue and peritransplantation rituximab. 253. Mueller-Eckhardt C, Salama A, Mahn I, Kiefel V, Neuzner J,
Blood 2002;99:1486–1488. Graubner M: Lack of efficacy of high-dose intravenous
229. Jourdan E, Topart D, Richard B, Jourdan J, Sotto A: Severe immunoglobulin in autoimmune haemolytic anaemia: A clue
autoimmune hemolytic anemia following rituximab therapy to its mechanism. Scand J Haematol 1985;34:394–400.
in a patient with a lymphoproliferative disorder. Leuk 254. Salama A, Mueller-Eckhardt C, Kiefel V: Effect of intravenous
Lymphoma 2003;44:889–890. immunoglobulin in immune thrombocytopenia. Lancet
230. Ahn YS, Harrington WJ, Mylvaganam R, Ayub J, Pall LM: 1983;2:193–195.
Danazol therapy for autoimmune hemolytic anemia. Ann 255. Flores G, Cunningham-Rundles C, Newland AC, Bussel JB:
Intern Med 1985;102:298–301. Efficacy of intravenous immunoglobulin in the treatment of
231. Ahn YS: Efficacy of danazol in hematologic disorders. Acta autoimmune hemolytic anemia: Results in 73 patients. Am J
Haematol 1990;84:122–129. Hematol 1993;44:237–242.
232. Manoharan A: Danazol therapy in patients with immune 256. Bussel JB, Cunningham-Rundles C, Abraham C: Intravenous
cytopenias. Aust N Z J Med 1987;17:613–614. treatment of autoimmune hemolytic anemia with very high
233. Tan AM, Lou J, Cheng HK: Danazol for treatment of refractory dose gammaglobulin. Vox Sang 1986;51:264–269.
autoimmune hemolytic anaemia. Ann Acad Med Singapore 257. Ratko TA, Burnett DA, Foulke GE, Matuszewski KA, Sacher
1989;18:707–709. RA: Recommendations for off-label use of intravenously
234. Pignon JM, Poirson E, Rochant H: Danazol in autoimmune administered immunoglobulin preparations. University
haemolytic anaemia. Br J Haematol 1993;83:343–345. Hospital Consortium Expert Panel for Off-Label Use of
235. Cervera H, Jara LJ, Pizarro S, Enkerlin HL, Fernandez M, Polyvalent Intravenously Administered Immunoglobulin
Medina F, et al: Danazol for systemic lupus erythematosus Preparations. JAMA 1995;273:1865–1870.
with refractory autoimmune thrombocytopenia or Evans’ 258. Ratko TA: Technology Assessment: Intravenous Immunoglo-
syndrome. J Rheumatol 1995;22:1867–1871. bulin Preparations. Oak Brook, IL, University HealthSystem
236. Chan AC, Sack K: Danazol therapy in autoimmune hemolytic Consortium, 1999.
anemia associated with systemic lupus erythematosus. 259. Sacher RA: Intravenous immunoglobulin consensus state-
J Rheumatol 1991;18:280–282. ment. J Allergy Clin Immunol 2001;108(4 Suppl.):S139–S146.
237. Lugassy G, Reitblatt T, Ducach A, Oren S: Severe autoimmune 260. Besa EC: Rapid transient reversal of anemia and long-term
hemolytic anemia with cold agglutinin and sclerodermic effects of maintenance intravenous immunoglobulin for
features—Favorable response to danazol. Ann Hematol autoimmune hemolytic anemia in patients with lymphopro-
1993;67:143–144. liferative disorders. Am J Med 1988;84:691–698.
238. Narayana SVL, Babu YS, Volanakis JE: Inhibition of 261. Leickly FE, Buckley RH: Successful treatment of autoimmune
Complement Serine Proteases as a Therapeutic Strategy. In hemolytic anemia in common variable immunodeficiency
Lambris JD, Holers VM (eda): Contemporary Immunology: with high-dose intravenous gamma globulin. Am J Med
Therapeutic Interventions in the Complement System. 1987;82:159–162.
Totowa, NJ: Homana Press, 2000:57–74. 262. Otheo E, Maldonado MS, Munoz A, Hernandez-Jodra M:
239. Quigg RJ: Modulation of disease using recombinant human High-dose intravenous immunoglobulin as single therapy in
endogenous complement inhibitors. In Lambris JD, Holers a child with autoimmune hemolytic anemia. Pediatr Hematol
VM (eds): Contemporary Immunology: Therapeutic Interven- Oncol 1997;14:487–490.
tions in the Complement System. Totowa, NJ: Homana Press, 263. Gonzalez CA: Successful treatment of autoimmune hemolytic
2000:155–170. anemia with intravenous immunoglobulin in a patient with
240. Walport MJ: Complement: First of two parts. N Engl J Med AIDS. Transplant Proc 1998;30:4151–4152.
2001;344:1058–1066. 264. Vandenberghe P, Zachee P, Verstraete S, Demuynck H,
241. Walport MJ: Complement: Second of two parts. N Engl J Med Boogaerts MA, Verhoef GE: Successful control of refractory
2001;344:1140–1144. and life-threatening autoimmune hemolytic anemia
242. Sahu A, Lambris JD: Complement inhibitors: A resurgent with intravenous immunoglobulins in a man with the
concept in anti-inflammatory therapeutics. Immunopharma- primary antiphospholipid syndrome. Ann Hematol 1996;
cology 2000;49:133–148. 73:253–256.
243. Kirschfink M: Targeting complement in therapy. Immunol 265. Argiolu F, Diana G, Arnone M, Batzella MG, Piras P, Cao A:
Rev 2001;180:177–189. High-dose intravenous immunoglobulin in the management
244. Basta M: Modulation of complement-mediated immune of autoimmune hemolytic anemia complicating thalassemia
damage by intravenous immune globulin. Clin Exp Immunol major. Acta Haematol 1990;83:65–68.
1996;104(Suppl. 1):21–25. 266. Salama A, Mahn I, Neuzner J, Graubner M, Mueller-Eckhardt
245. Yazdanbakhsh K, Kang S, Tamasauskas D, Sung D, C: IgG therapy in autoimmune haemolytic anaemia of warm
Scaradavou A: Complement receptor 1 inhibitors for preven- type. Blut 1984;48:391–392.
tion of immune-mediated red cell destruction: Potential use in 267. Atrah HI, Crawford RJ, Gabra GS, Mitchell R: Successes and
transfusion therapy. Blood 2003;101:5046–5052. failures of intravenous immunoglobulin. Scand J Haematol
246. Bjorkholm M: Intravenous immunoglobulin treatment in cyto- 1985;34:345–347.
penic haematological disorders. J Intern Med 1993;234:119–126. 268. American Society of Hospital Pharmacists (ASHP)
247. Kazatchkine MD, Kaveri SV: Immunomodulation of autoim- Commission of Therapeutics: ASHP therapeutic guidelines
mune and inflammatory diseases with intravenous immune for intravenous immune globulin. Clin Pharmacol 1992;11:
globulin. N Engl J Med 2001;345:747–755. 117–136.
248. Reilly MP, McKenzie SE: Mechanisms of action of IVIg: phy- 269. Schwartz SA: Clinical use of immune serum globulin as
siology of Fc receptors. Vox Sang 2002;83(Suppl. 1):57–63. replacement therapy in patients with primary immuno-
454 Immune Hemolytic Anemias

deficiency syndromes. In Ballow M (ed): IVIG Therapy Today. 292. Klaesson S, Ringden O, Ljungman P, Aschan J, Hagglund H,
Totowa, NJ: Humana Press, 1992:1–12. Winiarski J: Does high-dose intravenous immune globulin
270. Sherer Y, Levy Y, Langevitz P, Rauova L, Fabrizzi F, Shoenfeld treatment after bone marrow transplantation increase mortal-
Y: Adverse effects of intravenous immunoglobulin therapy in ity in veno-occlusive disease of the liver? Transplantation
56 patients with autoimmune diseases. Pharmacology 2001; 1995;60:1225–1230.
62:133–137. 293. Rizk A, Gorson KC, Kenney L, Weinstein R: Transfusion-
271. Sekul EA, Cupler EJ, Dalakas MC: Aseptic meningitis asso- related acute lung injury after the infusion of IVIG. Trans-
ciated with high-dose intravenous immunoglobulin therapy: fusion 2001;41:264–268.
Frequency and risk factors. Ann Intern Med 1994;121:259–262. 294. Isbister JP: Therapeutic apheresis. Indian J Pediatr
272. Perazella MA, Cayco AV: Acute renal failure and intravenous 2001;68:61–67.
immune globulin: Sucrose nephropathy in disguise? Am J 295. Grima KM: Therapeutic apheresis in hematological and onco-
Ther 1998;5:399–403. logical diseases. J Clin Apheresis 2000;15:28–52.
273. Friedman KD, Menitove JE: Preparation and Clinical Use of 296. Koo AP: Therapeutic apheresis in autoimmune and rheumatic
Plasma and Plasma Fractions. In Beutler E, Lichtman MA, diseases. J Clin Apheresis 2000;15:18–27.
Coller BS, Kipps TJ, Seligsohn U (eds): Williams Hematology. 297. Kiprov DD, Strauss RG, Ciavarella D, Gilcher RO, Kasprisin
New York: McGraw-Hill, 2001:1917–1934. DO, Klein HG, et al: Management of autoimmune disorders.
274. Epstein JS, Zoon KC: Important drug warning. FDA Dear J Clin Apheresis 1993;8:195–210.
Doctor Letter. Nov. 13, 1998. Rockville, MD: Center for 298. McLeod BC, Strauss RG, Ciavarella D, Gilcher RO, Kasprisin
Biologics Evalulation and Research, Food and Drug DO, Kiprov DD, et al: Management of hematological disor-
Administration. ders and cancer. J Clin Apheresis 1993;8:211–230.
275. Stahl M, Schifferli JA: The renal risks of high-dose intravenous 299. Branda RF, Moldow CF, McCullough JJ, Jacob HS: Plasma
immunoglobulin treatment. Nephrol Dial Transplant exchange in the treatment of immune disease. Transfusion
1998;13:2182–2185. 1975;15:570–576.
276. Haskin JA, Warner DJ, Blank DU: Acute renal failure after 300. Patten E, Reuter FP: Evans’ syndrome: Possible benefit from
large doses of intravenous immune globulin. Ann Pharma- plasma exchange. Transfusion 1980;20:589–593.
cother 1999;33:800–803. 301. Rosenfield RE, Jagathambal: Transfusion therapy for autoim-
277. Gaines A, Varricchio F, Kapit R, Pierce LR, Scott D, Finlayson mune hemolytic anemia. Semin Hematol 1976;13:311–321.
J: Renal insufficiency and failure associated with immune 302. Garelli S, Mosconi L, Valbonesi M, Schieppati G, Navassa G:
globulin intravenous therapy-United States (1985–98). Morb Plasma exchange for a hemolytic crisis due to autoimmune
Mort Weekly Rpt 1999;48:518–521. hemolytic anemia of the IgG warm type. Blut 1980;41:387–391.
278. Ahsan N: Intravenous immunoglobulin induced-nephropathy: 303. Bernstein ML, Schneider BK, Naiman JL: Plasma exchange in
A complication of IVIG therapy. J Nephrol 1998;11:157–161. refractory acute autoimmune hemolytic anemia. J Pediatr
279. Laidlaw S, Bainton R, Wilkie M, Makris M: Acute renal failure 1981;98:774–775.
in acquired haemophilia following the use of high dose intra- 304. Schroeder JO, Euler HH, Loffler H: Synchronization of plasma-
venous immunoglobulin. Haemophilia 1999;5:270–272. pheresis and pulse cyclophosphamide in severe systemic lupus
280. Levy JB, Pusey CD: Nephrotoxicity of intravenous immuno- erythematosus. Ann Intern Med 1987;107:344–346.
globulin. QJM 2000;93:751–755. 305. Hughes P, Toogood A: Plasma exchange as a necessary pre-
281. Sati HI, Ahya R, Watson HG: Incidence and associations of requisite for the induction of remission by human
acute renal failure complicating high-dose intravenous immunoglobulin in auto-immune haemolytic anaemia. Acta
immunoglobulin therapy. Br J Haematol 2001;113:556–557. Haematol 1994;91:166–169.
282. Gupta N, Ahmed I, Nissel-Horowitz S, Patel D, Mehrotra B: 306. Silberstein LE, Berkman EM: Plasma exchange in autoimmune
Intravenous gammglobulin-associated acute renal failure. Am hemolytic anemia (AIHA). J Clin Apheresis 1983;1:238–242.
J Hematol 2001;66:151–152. 307. Brooks BD, Steane EA, Sheehan RG, Frenkel EP: Therapeutic
283. Go RS, Call TG: Deep venous thrombosis of the arm after plasma exchange in the immune hemolytic anemias and
intravenous immunoglobulin infusion: Case report and litera- immunologic thrombocytopenic purpura. Prog Clin Biol Res
ture review of intravenous immunoglobulin-related throm- 1982;106:317–329.
botic complications. Mayo Clin Proc 2000;75:83–85. 308. Geurs F, Ritter K, Mast A, Van Maele V: Successful plasma-
284. Dalakas MC: High-dose intravenous immunoglobulin and pheresis in corticosteroid-resistant hemolysis in infectious
serum viscosity: Risk of precipitating thromboembolic events. mononucleosis: Role of autoantibodies against triosephos-
Neurology 1994;44:223–226. phate isomerase. Acta Haematol 1992;88:142–146.
285. Woodruff RK, Grigg AP, Firkin FC, Smith IL: Fatal thrombotic 309. Kutti J, Wadenvik H, Safai-Kutti S, Bjorkander J, Hanson LA,
events during treatment of autoimmune thrombocytopenia Westberg G, et al: Successful treatment of refractory autoim-
with intravenous immunoglobulin in elderly patients. Lancet mune haemolytic anaemia by plasmapheresis. Scand J
1986;2:217–218. Haematol 1984;32:149–152.
286. Silbert PL, Knezevic WV, Bridge DT: Cerebral infarction com- 310. Andersen O, Taaning E, Rosenkvist J, Moller NE, Mogensen
plicating intravenous immunoglobulin therapy for polyneuri- HH: Autoimmune haemolytic anaemia treated with multiple
tis cranialis. Neurology 1992;42:257–258. transfusions, immunosuppressive therapy, plasma exchange,
287. Reinhart WH, Berchtold PE: Effect of high-dose intravenous and desferrioxamine. Acta Paediatr Scand 1984;73:145–148.
immunoglobulin therapy on blood rheology. Lancet 311. von Keyserlingk H, Meyer-Sabellek W, Arntz R, Haller H:
1992;339:662–664. Plasma exchange treatment in autoimmune hemolytic anemia
288. Steg RE, Lefkowitz DM: Cerebral infarction following intra- of the warm antibody type with renal failure. Vox Sang
venous immunoglobulin therapy for myasthenia gravis. 1987;52:298–300.
Neurology 1994;44:1180–1181. 312. McConnell ME, Atchison JA, Kohaut E, Castleberry RP:
289. Brannagan TH, III, Nagle KJ, Lange DJ, Rowland LP: Successful use of plasma exchange in a child with refractory
Complications of intravenous immune globulin treatment in immune hemolytic anemia. Am J Pediatr Hematol Oncol
neurologic disease. Neurology 1996;47:674–677. 1987;9:158–160.
290. Fisman DN, Smilovitch M: Intravenous immunoglobulin, 313. McCarthy LJ, Danielson CF, Fernandez C, Skipworth E,
blood viscosity and myocardial infarction. Can J Cardiol Limiac CA, Prahlow T, et al: Intensive plasma exchange for
1997;13:775–777. severe autoimmune hemolytic anemia in a four-month-old
291. Wolff SN, Fay JW, Herzig RH, et al: High-dose weekly intra- infant. J Clin Apheresis 1999;14:190–192.
venous immunoglobulin to prevent infections in patients 314. Shehata N, Kouroukis C, Kelton JG: A review of randomized
undergoing autologous bone marrow transplantation or controlled trials using therapeutic apheresis. Transfus Med
severe myelosuppressive therapy. A study of the American Rev 2002;16:200–229.
Bone Marrow Transplant Group. Ann Intern Med 1993;118: 315. Smith JW, Weinstein R, for The AABB Hemapheresis
937–942. Committee KL: Therapeutic apheresis: A summary of current
Management of Autoimmune Hemolytic Anemias 455

indication categories endorsed by the AABB and the 337. Burt RK, Traynor AE: Hematopoietic stem cell transplanta-
American Society for Apheresis. Transfusion 2003;43:820–822. tion: A new therapy for autoimmune disease. Stem Cells
316. Cash JM, Wilder RL: Treatment-resistant rheumatic disease. 1999;17:366–372.
Rheum Dis Clin N Am 1995;21:1–170. 338. Huhn RD, Fogarty PF, Nakamura R, et al: High-dose cyclopho-
317. Lafferty KL, Gazda KS: Costimulation and the regulation of sphamide with autologous lymphocyte-depleted peripheral
autoimmunity. In Rose NR, Mackay IR (eds): The Autoimmune blood stem cell (PBSC) support for treatment of refractory
Diseases. San Diego, CA: Academic Press, 1998:59–74. chronic autoimmune thrombocytopenia. Blood 2003;101:71–77.
318. Bingham SJ, Snowden J, Morgan G, Emery P: High dose 339. Paillard C, Kanold J, Halle P, et al: Two-step immunoablative
immunosuppressive therapy and stem cell transplantation in treatment with autologous peripheral blood CD34(+) cell
autoimmune and inflammatory diseases. Int Immuno- transplantation in an 8–year-old boy with autoimmune
pharmacol 2002;2:399–414. haemolytic anaemia. Br J Haematol 2000;110:900–902.
319. Marmont AM: New horizons in the treatment of autoimmune 340. Burt RK, Schroeder J, Rosa R, et al: Autologous hematopoietic
diseases: Immunoablation and stem cell transplantation. stem cell transplantation of patients with severe and refrac-
Annu Rev Med 2000;51:115–134. tory systemic lupus erythematosus (SLE): Three year follow-
320. Van Bekkum DW: Autologous stem cell transplantation for up [abstract]. Blood 2000;96(Suppl. 1):843.
treatment of autoimmune diseases. Stem Cells 1999;17:172–178. 341. Musso M, Porretto F, Crescimanno A, et al: Autologous
321. Moore J, Brooks P: Stem cell transplantation for autoimmune peripheral blood stem and progenitor (CD34+) cell transplan-
diseases. Springer Semin Immunopathol 2001;23:193–213. tation for systemic lupus erythematosus complicated by
322. Cohen Y, Polliack A, Nagler A: Treatment of refractory Evans syndrome. Lupus 1998;7:492–494.
autoimmune diseases with ablative immunotherapy using 342. Snowden JA, Patton WN, O’Donnell JL, Hannah EE, Hart DN:
monoclonal antibodies and/or high dose chemotherapy with Prolonged remission of longstanding systemic lupus
hematopoietic stem cell support. Curr Pharm Des erythematosus after autologous bone marrow transplant for
2003;9:279–288. non-Hodgkin’s lymphoma. Bone Marrow Transplant
323. Van Bekkum DW: Experimental basis of hematopoietic stem 1997;19:1247–1250.
cell transplantation for treatment of autoimmune diseases. J 343. Rosler W, Manger B, Repp R, Kalden JR, Gramatzki M: Auto-
Leukoc Biol 2002;72:609–620. logous PBPCT in a patient with lymphoma and Sjogren’s syn-
324. Burt RK, Slavin S, Burns WH, Marmont AM: Induction of drome: complete remission of lymphoma without control of the
tolerance in autoimmune diseases by hematopoietic stem cell autoimmune disease. Bone Marrow Transplant 1998;22:211–213.
transplantation: Getting closer to a cure? Blood 2002;99: 344. Brodsky RA, Sensenbrenner LL, Jones RJ: Complete remission
768–784. in severe aplastic anemia after high-dose cyclophosphamide
325. Morton JL, Siegel BV: Transplantation of autoimmune poten- without bone marrow transplantation. Blood 1996;87:491–494.
tial. Development of antinuclear antibodies in H-2 histocom- 345. Brodsky RA, Petri M, Smith BD, Seifter EJ, Spivak JL, Styler
patible recipients of bone marrow from New Zealand Black M, et al: Immunoablative high-dose cyclophosphamide
mice. Proc Natl Acad Sci U S A 1974;71:2162–2166. without stem-cell rescue for refractory, severe autoimmune
326. Ikehara S: Autoimmune diseases as stem cell disorders: disease. Ann Intern Med 1998;129:1031–1035.
Normal stem cell transplant for their treatment [Review]. Int J 346. Brodsky RA, Smith BD: Bone marrow transplantation for
Mol Med 1998;1:5–16. autoimmune diseases. Curr Opin Oncol 1999;11:83–86.
327. Ikehara S: Bone marrow transplantation for autoimmune dis- 347. Nousari HC, Brodsky RA, Jones RJ, Grever MR, Anhalt GJ:
eases. Acta Haematol 1998;99:116–132. Immunoablative high-dose cyclophosphamide without stem
328. Li H, Kaufman CL, Boggs SS, Johnson PC, Patrene KD, Ildstad cell rescue in paraneoplastic pemphigus: Report of a case and
ST: Mixed allogeneic chimerism induced by a sublethal review of this new therapy for severe autoimmune disease.
approach prevents autoimmune diabetes and reverses insuli- J Am Acad Dermatol 1999;40(5 Pt 1):750–754.
tis in nonobese diabetic (NOD) mice. J Immunol 1996;156: 348. Marmont AM: Immunoablation followed or not by
380–388. hematopoietic stem cells as an intense therapy for severe
329. Van Bekkum DW: BMT in experimental autoimmune dis- autoimmune diseases. New perspectives, new problems.
eases. Bone Marrow Transplant 1993;11:183–187. Haematologica 2001;86:337–345.
330. Raetz E, Beatty PG, Adams RH: Treatment of severe Evans 349. Euler HH, Schroeder JO, Harten P, Zeuner RA, Gutschmidt
syndrome with an allogeneic cord blood transplant. Bone HJ: Treatment-free remission in severe systemic lupus erythe-
Marrow Transplant 1997;20:427–429. matosus following synchronization of plasmapheresis with
331. De Stefano P, Zecca M, Giorgiani G, Perotti C, Giraldi E, subsequent pulse cyclophosphamide. Arthritis Rheum
Locatelli F: Resolution of immune haemolytic anaemia with 1994;37:1784–1794.
allogeneic bone marrow transplantation after an unsuccessful 350. Mielcarek M, Sandmaier BM, Maloney DG, Maris M,
autograft. Br J Haematol 1999;106:1063–1064. McSweeney PA, Woolfrey A, et al: Nonmyeloablative hemato-
331a. Oyama Y, Papadopoulos EB, Miranda M, Traynor AE, Burt poietic cell transplantation: Status quo and future perspec-
RK: Allogeneic stem cell transplantation for Evans syndrome. tives. J Clin Immunol 2002;22:70–74.
Bone Marrow Transplant 2001;28:903–905. 351. Champlin R, Khouri I, Shimoni A, Gajewski J, Kornblau S,
332. Marmont AM, Gualandi F, Van Lint MT, Bacigalupo A: Molldrem J, et al: Harnessing graft-versus-malignancy: Non-
Refractory Evans’ syndrome treated with allogeneic SCT fol- myeloablative preparative regimens for allogeneic haemato-
lowed by DLI: Demonstration of a graft-versus-autoimmunity poietic transplantation, an evolving strategy for adoptive
effect. Bone Marrow Transplant 2003;31:399–402. immunotherapy. Br J Haematol 2000;111:18–29.
333. Hinterberger W, Hinterberger-Fischer M, Marmont A: 352. Barrett J, Childs R: Non-myeloablative stem cell transplants.
Clinically demonstrable anti-autoimmunity mediated by allo- Br J Haematol 2000;111:6–17.
geneic immune cells favorably affects outcome after stem cell 353. Pulsipher MA, Woolfrey A: Nonmyeloablative transplanta-
transplantation in human autoimmune diseases. Bone tion in children: Current status and future prospects. Hematol
Marrow Transplant 2002;30:753–759. Oncol Clin North Am 2001;15:809–834.
334. Burt RK, Traynor AE, Craig R, Marmont AM: The promise of 354. Baron F, Beguin Y: Nonmyeloablative allogeneic hemato-
hematopoietic stem cell transplantation for autoimmune dis- poietic stem cell transplantation. J Hematother Stem Cell Res
eases. Bone Marrow Transplant 2003;31:521–524. 2002;11:243–263.
335. Fassas A: Intense immunosuppression and autologous 355. Wilmers MJ, Russell PA: Autoimmune hemolytic anemia in an
hematopoietic stem cell transplantation for multiple sclerosis. infant treated by thymectomy. Lancet 1963;2:915.
Haematologica 2003;88:244–245. 356. Hancock DM: Autoimmune haemolytic anaemia in an infant
336. Openshaw H, Nash RA, McSweeney PA: High-dose immuno- treated by thymectomy. Lancet 1963;2:1118.
suppression and hematopoietic stem cell transplantation in 357. Karaklis AS, Valaes T, Pantelakis SN, Doxiadis SA:
autoimmune disease: Clinical review. Biol Blood Marrow Thymectomy in an infant with autoimmune haemollytic
Transplant 2002;8:233–248. anaemia. Lancet 1964;2:778.
456 Immune Hemolytic Anemias

358. Oski FA, Abelson NM: Autoimmune hemolytic anemia in an 381. Schubothe H: The cold hemagglutinin disease. Semin
infant: Report of a case treated unsuccessfully with thymec- Hematol 1966;3:27–47.
tomy. J Pediatr 1965;67:752–758. 382. Worlledge SM, Brain MC, Cooper AC, Hobbs JR, Dacie JV:
359. Hirooka M, Yoshioka K, Ono T, Kubota N, Ikeda S: Immmunosuppressive drugs in the treatment of autoimmune
Autoimmune hemolytic anemia in a child treated with haemolytic anaemia. Proc R Soc Med 1968;61:1312–1315.
thymectomy. Tohoku J Exp Med 1970;101:227–235. 383. Hippe E, Jensen KB, Olesen H, Lind K, Thomsen PE:
360. Rennenberg RJ, Pauwels P, Vlasveld LT: A case of thymoma- Chlorambucil treatment of patients with cold agglutinin
associated autoimmune haemolytic anaemia. Neth J Med syndrome. Blood 1970;35:68–72.
1997;50:110–114. 384. Olesen H: Chlorambucil treatment in the cold agglutinin
361. Albahary C, Homberg JC, Guillaume J, Martin S, Boulangiez syndrome. Scand J Haematol 1964;1:116.
JP: Thymoma, myasthenia and auto-immune hemolytic 385. Evans RS, Baxter E, Gilliland BC: Chronic hemolytic anemia
anemia [in French]. Nouv Presse Med 1972;1:1931–1934. due to cold agglutinins: A 20-year history of benign gammo-
362. Arntzenius AB, Bieger R: Disappearance of autoantibody- pathy with response to chlorambucil. Blood 1973;42:463–470.
induced haemolysis after excision of a malignant thymoma. 386. Ramos RR, Curtis BR, Sadler JE, Eby CS, Chaplin H:
Neth J Med 1991;38:117–121. Refractory immune hemolytic anemia with a high thermal
363. Dreyfus B, Aubert P, Patte D, Bolloc’h-Combrisson Le A: amplitude, low affinity IgG anti-Pra cold autoantibody.
Erythroblastopenic chronique decouverte apres une thymec- Autoimmunity 1992;12:149–154.
tomie. Nouv Rev Fr Hematol 1963;3:765–772. 387. Chaplin H Jr: Lymphoma in primary chronic cold hemagglu-
364. Fischer JT, Lautenschlager J, Pottgen W: Autoimmune tinin disease treated with chlorambucil. Arch Intern Med
haemolytic anaemia following malignant thymoma [in 1982;142:2119–2123.
German]. Dtsch Med Wochenschr 1974;99:1867–1869. 388. Jacobs A: Cold agglutinin hemolysis responding to fludara-
365. Halperin IC, Minogue WF, Komninos ZD: Autoimmune bine therapy. Am J Hematol 1996;53:279–280.
hemolytic anemia and myasthenia gravis associated with 389. Di Raimondo F, Giustolisi R, Cacciola E, O’Brien S, Kantarjian
thymoma. N Engl J Med 1966;275:663–664. H, Robertson LB, et al: Autoimmune hemolytic anemia in
365a. Hennemann HH, Beck T: Autoimmunohaemolytische Anamie chronic lymphocytic leukemia patients treated with fludara-
nach Bestrahlung eines Thymoma. Dtsch Med Wochenschr bine. Leuk Lymphoma 1993;11:63–68.
1974;99:1869–1871. 390. Berentsen S, Tjonnfjord GE, Brudevold R, Gjertsen BT,
366. Janbon MM, Bertrand L, Bonnet H: Sarcome du thymus avec Langholm R, Lokkevik E, et al: Favourable response to
anemie hemolytique par auto-anticorps. Montpellier Med therapy with the anti-CD20 monoclonal antibody rituximab
1956;49:161–164. in primary chronic cold agglutinin disease. Br J Haematol
367. Mongan ES, Kern WA Jr, Terry R: Hypogammaglobulinemia 2001;115:79–83.
with thymoma, hemolytic anemia, and disseminated infection 391. Pulik M, Genet P, Lionnet F, Touahri T: Treatment of primary
with cytomegalovirus. Ann Intern Med 1966;65:548–554. chronic cold agglutinin disease with rituximab: Maintenance
368. Pirofsky B: Autoimmune hemolytic anemia and neoplasia of therapy may improve the results. Br J Haematol 2002;117:
the reticuloendothelium: With a hypothesis concerning etio- 998–999.
logic relationships. Ann Intern Med 1968;68:109–121. 392. Lee EJ, Kueck B: Rituxan in the treatment of cold agglutinin
369. Ross JF, Finch SC, Street RB, Streider JW: The simultaneous disease. Blood 1998;92:3490–3491.
occurrence of benign thymoma and refractory anemia. Blood 393. Bauduer F: Rituximab: A very efficient therapy in cold
1954;9:935. agglutinins and refractory autoimmune haemolytic anaemia
370. Rubinstein I, Langevitz P, Hirsch R, Berkowicz M, Lieberman associated with CD20-positive, low-grade non-Hodgkin’s
Y, Shibi G: Autoimmune hemolytic anemia as the presenting lymphoma. Br J Haematol 2001;112:1085–1086.
manifestation of malignant thymoma. Acta Haematol 1985; 394. Layios N, Van Den NE, Jost E, Deneys V, Scheiff JM, Ferrant A:
74:40–42. Remission of severe cold agglutinin disease after Rituximab
371. Taniguchi S, Shibuya T, Morioka E, Okamura T, Okamura S, therapy. Leukemia 2001;15:187–188.
Inaba S, et al: Demonstration of three distinct immunological 395. Gharib M, Poynton C: Complete, long-term remission of
disorders on erythropoiesis in a patient with pure red cell refractory idiopathic cold haemagglutinin disease after
aplasia and autoimmune haemolytic anaemia associated with Mabthera. Br J Haematol 2002;117:248–249.
thymoma. Br J Haematol 1988;68:473–477. 396. Zaja F, Russo D, Fuga G, Michelutti T, Sperotto A, Fanin R,
372. Tiber C, Casimir M, Nogeire C, Lichtiger B, Conrad FG: et al: Rituximab in a case of cold agglutinin disease. Br J
Thymoma with red cell aplasia and hemolytic anemia. South Haematol 2001;115:232–233.
Med J 1981;74:1164–1165. 397. Engelhardt M, Jakob A, Ruter B, Trepel M, Hirsch F, Lubbert
373. Lyckholm LJ, Edmond MB: Images in clinical medicine. M: Severe cold hemagglutinin disease (CHD) successfully
Seasonal hemolysis due to cold-agglutinin syndrome. N Engl treated with rituximab. Blood 2002;100:1922–1923.
J Med 1996;334:437. 398. Silberstein LE, Robertson GA, Harris AC, Moreau L, Besa E,
374. Pisciotta AV: Cold hemagglutination in acute and chronic Nowell PC: Etiologic aspects of cold agglutinin disease:
hemolytic syndromes. Blood 1955;10:295. Evidence for cytogenetically defined clones of lymphoid cells
375. Firkin BG, Blackwell JB, Johnston GAW: Essential cryoglobu- and the demonstration that an anti-Pr cold autoantibody is
linaemia and acquired haemolytic anaemia due to cold agglu- derived from a chromosomally aberrant B cell clone. Blood
tinins. Australian Ann Med 1959;8:151. 1986;67:1705–1709.
376. Schreiber AD, Herskovitz BS, Goldwein M: Low-titer cold- 399. O’Connor BM, Clifford JS, Lawrence WD, Logue GL: Alpha-
hemagglutinin disease: Mechanism of hemolysis and interferon for severe cold agglutinin disease. Ann Intern Med
response to corticosteroids. N Engl J Med 1977;296:1490–1494. 1989;111:255–256.
377. Silberstein LE, Berkman EM, Schreiber AD: Cold hemagglu- 400. Fest T, de Wazieres B, Lamy B, Maskani M, Vuitton D,
tinin disease associated with IgG cold-reactive antibody. Ann Dupond JL: Successful response to alpha-interferon 2b in a
Intern Med 1987;106:238–242. refractory IgM autoagglutinin-mediated hemolytic anemia.
378. Dellagi K, Brouet JC, Schenmetzler C, Praloran V: Chronic Ann Hematol 1994;69:147–149.
hemolytic anemia due to a monoclonal IgG cold agglutinin 401. Isbister JP, Biggs JC, Penny R: Experience with large volume
with anti-Pr specificity. Blood 1981;57:189–191. plasmapheresis in malignant paraproteinaemia and immune
379. Nanan R, Scheurlen W, Gerlich M, Huppertz HI: Severe low- disorders. Aust N Z J Med 1978;8:154–164.
titer cold-hemagglutinin disease responsive to steroid pulse 402. Wells JV, Fudenberg HH: Paraproteinemias. Dis Mon 1974;1–45.
therapy. Ann Hematol 1995;71:101–102. 403. Logue GL, Rosse WF, Gockerman JP: Measurement of the
380. Lahav M, Rosenberg I, Wysenbeek AJ: Steroid-responsive third component of complement bound to red blood cells in
idiopathic cold agglutinin disease: A case report. Acta patients with the cold agglutinin syndrome. J Clin Invest
Haematol 1989;81:166–168. 1973;52:493.
Management of Autoimmune Hemolytic Anemias 457

404. Buskard NA: Plasma exchange and plasmapheresis. Can Med lymphocytic leukaemia associated with autoimmune haemo-
Assoc J 1978;119:681–683. lysis. Acta Haematol Pol 1994;25:119–127.
405. Rodenhuis S, Maas A, Hazenberg CA, Das PC, Nieweg HO: 425. Coad JE, Matutes E, Catovsky D: Splenectomy in lymphopro-
Inefficacy of plasma exchange in cold agglutinin hemolytic liferative disorders: A report on 70 cases and review of the
anemia—A case study. Vox Sang 1985;49:20–25. literature. Leuk Lymphoma 1993;10:245–264.
406. Valbonesi M, Guzzini F, Zerbi D, Villa P, Montani F, Angelini 426. Gentile TC, Loughran TP Jr: Resolution of autoimmune hemo-
G: Successful plasma exchange for a patient with chronic lytic anemia following splenectomy in CD3+ large granular
demyelinating polyneuropathy and cold agglutinin disease lymphocyte leukemia. Leuk Lymphoma 1996;23:405–408.
due to anti-Pra. J Clin Apheresis 1986;3:109–110. 427. Cusack JC Jr, Seymour JF, Lerner S, Keating MJ, Pollock RE:
406a. Bell CA, Zwicker H, Sacks HJ: Autoimmune hemolytic Role of splenectomy in chronic lymphocytic leukemia. J Am
anemia: Routine serologic evaluation in a general hospital Coll Surg 1997;185:237–243.
population. Am J Clin Pathol 1973;60:903–911. 428. Seymour JF, Cusack JD, Lerner SA, Pollock RE, Keating MJ:
407. von dem Borne AE, Engelfriet CP, Beckers D, Kort-Henkes G, Case/control study of the role of splenectomy in chronic lym-
Van Der GM, Van Loghem JJ: Autoimmune haemolytic phocytic leukemia. J Clin Oncol 1997;15:52–60.
anaemias. II. Warm haemolysins—Serological and immuno- 429. Neal TF Jr, Tefferi A, Witzig TE, Su J, Phyliky RL, Nagorney
chemical investigations and 51Cr studies. Clin Exp Immunol DM: Splenectomy in advanced chronic lymphocytic leukemia:
1969;4:333–343. a single institution experience with 50 patients. Am J Med
408. Evans RS, Bingham M, Turner E: Autoimmune hemolytic 1992;93:435–440.
disease: Observations of serologic reactions and disease 430. Besa EC: Use of intravenous immunoglobulin in chronic lym-
activity. Ann NY Acad Sci 1965;124:422. phocytic leukemia. Am J Med 1984;76:209–218.
409. Evans RS, Turner E, Bingham M, Woods R: Chronic hemolytic 431. Sigler E, Shtalrid M, Goland S, Sthoeger ZM, Berrebi A: Intract-
anemia due to cold agglutinins. II. The role of C’ in red cell able acute autoimmune hemolytic anemia in B-cell chronic
destruction. J Clin Invest 1968;47:691–701. lymphocytic leukemia successfully treated with vincristine-
410. Geffray E, Najman A: Efficacy of danazol in autoimmune loaded platelet infusion. Am J Hematol 1995;50:313–315.
hemolytic anemia with cold agglutinins: 4 cases. Presse Med 432. Willis F, Marsh JC, Bevan DH, Killick SB, Lucas G, Griffiths R,
1992;21:1472–1475. et al: The effect of treatment with Campath-1H in patients with
411. Ramos RR, Curtis BR, Eby CS, Ratkin GA, Chaplin H: Fatal autoimmune cytopenias. Br J Haematol 2001;114:891–898.
outcome in a patient with autoimmune hemolytic anemia 433. Rodon P, Breton P, Courouble G: Treatment of pure red cell
associated with an IgM bithermic anti-ITP. Transfusion aplasia and autoimmune haemolytic anaemia in chronic lym-
1994;34:427–431. phocytic leukaemia with Campath-1H. Eur J Haematol
412. Seldon M, Isbister JP, Raik E, Biggs JC: A fatal case of cold 2003;70:319–321.
autoimmune hemolytic anemia. Am J Clin Pathol 1980;73: 434. Gupta N, Kavuru S, Patel D, Janson D, Driscoll N, Ahmed S,
716–717. et al: Rituximab-based chemotherapy for steroid-refractory
413. Rousey SR, Smith RE: A fatal case of low titer anti-PR cold autoimmune hemolytic anemia of chronic lymphocytic
agglutinin disease. Am J Hematol 1990;35:286–287. leukemia. Leukemia 2002;16:2092–2095.
414. Hillen HF, Bakker SJ: Failure of interferon-alpha-2b therapy in 435. Zaja F, Iacona I, Masolini P, Russo D, Sperotto A, Prosdocimo
chronic cold agglutinin disease. Eur J Haematol 1994;53:242–243. S, et al: B-cell depletion with rituximab as treatment for
415. Berentsen S, Tjonnfjord GE, Shammas FV, Bergheim J, immune hemolytic anemia and chronic thrombocytopenia.
Hammerstrom J, Langholm R, et al: No response to cladribine Haematologica 2002;87:189–195.
in five patients with chronic cold agglutinin disease. Eur J 436. Kokori SI, Ioannidis JP, Voulgarelis M, Tzioufas AG,
Haematol 2000;65:88–90. Moutsopoulos HM: Autoimmune hemolytic anemia in
416. Gottsche B, Salama A, Mueller-Eckhardt C: Donath- patients with systemic lupus erythematosus. Am J Med
Landsteiner autoimmune hemolytic anemia in children: A 2000;108:198–204.
study of 22 cases. Vox Sang 1990;58:281–286. 437. Voulgarelis M, Kokori SI, Ioannidis JP, Tzioufas AG, Kyriaki
417. Banov CH: Paroxysmal cold hemoglobinuria: Apparent D, Moutsopoulos HM: Anaemia in systemic lupus erythe-
remission after splenectomy. JAMA 1960;174:1974. matosus: Aetiological profile and the role of erythropoietin.
418. Roy-Burman A, Glader BE: Resolution of severe Donath- Ann Rheum Dis 2000;59:217–222.
Landsteiner autoimmune hemolytic anemia temporally asso- 438. Harvey AM, Shulman LE, Tumulty PA, et al: Systemic lupus
ciated with institution of plasmapheresis. Crit Care Med erythematosus. Review of the literature and clinical analysis
2002;30:931–934. of 138 cases. Medicine 1954;33:291–437.
418a. Ries CA, Garratty G, Petz LD, Fudenberg HH: Paroxysmal 439. Salamon DJ, Ramsey G, Nusbacher J, Yang S, Starzl TE, Israel
cold hemoglobinuria: Report of a case with an exceptionally L: Anti-A production by a group O spleen transplanted to a
high thermal range Donath-Landsteiner antibody. Blood group A recipient. Vox Sang 1985;48:309–312.
1971;38:491–499. 440. Alger M, Alarcon-Segovia D, Rivero SJ: Hemolytic anemia
419. Mauro FR, Foa R, Cerretti R, Giannarelli D, Coluzzi S, and thrombocytopenic purpura: Two related subsets of
Mandelli F, et al: Autoimmune hemolytic anemia in chronic systemic lupus erythematosus. J Rheumatol 1977;4:351–357.
lymphocytic leukemia: clinical, therapeutic, and prognostic 441. Ward MM, Pyun E, Studenski S: Mortality risks associated
features. Blood 2000;95:2786–2792. with specific clinical manifestations of systemic lupus erythe-
420. Cheson BD, Bennett JM, Rai KR, Grever MR, Kay NE, Schiffer matosus. Arch Intern Med 1996;156:1337–1344.
CA, et al: Guidelines for clinical protocols for chronic 442. Alarcon-Segovia D, Deleze M, Oria CV, Sanchez-Guerrero J,
lymphocytic leukemia: Recommendations of the National Gomez-Pacheco L, Cabiedes J, et al: Antiphospholipid anti-
Cancer Institute-sponsored working group. Am J Hematol bodies and the antiphospholipid syndrome in systemic lupus
1988;29:152–163. erythematosus. A prospective analysis of 500 consecutive
421. McCann EL, Shirey RS, Kickler TS, Ness PM: IgM auto- patients. Medicine (Baltimore) 1989;68:353–365.
agglutinins in warm autoimmune hemolytic anemia: A poor 443. Deleze M, Alarcon-Segovia D, Oria CV, Sanchez-Guerrero J,
prognostic feature. Acta Haematol 1992;88:120–125. Fernandez-Dominguez L, Gomez-Pacheco L, et al: Hemocyto-
422. Pisciotta AV, Hirschboeck JS: Therapeutic considerations in penia in systemic lupus erythematosus. Relationship to
chronic lymphocytic leukemia. Arch Intern Med 1957;99: antiphospholipid antibodies. J Rheumatol 1989;16:926–930.
334–345. 444. McDonagh JE, Isenberg DA: Development of additional
423. Piro LD, Carrera CJ, Beutler E, Carson DA: 2–Chlorode- autoimmune diseases in a population of patients with sys-
oxyadenosine: An effective new agent for the treatment of temic lupus erythematosus. Ann Rheum Dis 2000;59:230–232.
chronic lymphocytic leukemia. Blood 1988;72:1069–1073. 445. Sarles HE, Levin WC: The role of splenectomy in the manage-
424. Zaucha JM, Halaburda K, Ciepluch H, Hellmann A: 2- ment of acquired autoimmune hemolytic anemia complicat-
Chlorodeoxyadenosine treatment of patients with chronic ing systemic lupus erythematosus. Amer J Med 1959;26:547.
458 Immune Hemolytic Anemias

446. Dubois EL: Systemic lupus erythematosus. M Clin North Am 471. Sokol RJ, Booker DJ, Stamps R: Erythrocyte autoantibodies,
1952;36:1111. autoimmune haemolysis, and carcinoma. J Clin Pathol
447. Haserick JR: Modern concepts of systemic lupus erythemato- 1994;47:340–343.
sus; a review of 126 cases. J Chronic Dis 1955;1:317. 472. Gordon PA, Baylis PH, Bird GW: Tumour-induced autoim-
448. Johnson HM: The effect of splenectomy in acute systemic mune haemolytic anaemia. Br Med J 1976;1:1569–1570.
lupus erythematosus. Arch Dermatol Syph 1953;68:699. 473. Horne MK, McAnally TP: Hemolytic anemia with lung carci-
449. Pisciotta AV, Giliberti JJ, Greenwalt TJ, Engstrom WW: Acute noma: Case reports. Milit Med 1978;143:188–189.
hemolytic anemia in disseminated lupus erythematosus. 474. Carreras Vescio LA, Toblli JE, Rey JA, Assaf ME, De Maria HE,
Amer J Clin Path 1951;21:1139. Marletta J: Autoimmune hemolytic anemia associated with an
450. Gruenberg JC, VanSlyck EJ, Abraham JP: Splenectomy in sys- ovarian neoplasm. Medicina (B Aires) 1983;43:415–424.
temic lupus erythematosis. Am Surg 1986;52:366–370. 475. Girelli G, Adorno G, Perrone MP, et al: Kidney carcinoma
451. Rivero SJ, Alger M, Alarcon-Segovia D: Splenectomy for revealed by autoimmune hemolytic anemia. Haematologica
hemocytopenia in systemic lupus erythematosus: A control- 1988;73:309–311.
led appraisal. Arch Intern Med 1979;139:773–776. 476. Adorno G, Girelli G, Perrone MP, et al: A metastatic breast car-
452. Drenkard C, Villa AR, Alarcon-Segovia D, Perez-Vazquez ME: cinoma presenting as autoimmune hemolytic anemia. Tumori
Influence of the antiphospholipid syndrome in the survival of 1991;77:447–448.
patients with systemic lupus erythematosus. J Rheumatol 477. Hibino S, Stoller RG, Jacobs SA: Autoimmune haemolytic
1994;21:1067–1072. anaemia associated with transitional cell carcinoma. Lancet
453. Nossent JC, Swaak AJ: Prevalence and significance of haema- 1992;340:373.
tological abnormalities in patients with systemic lupus ery- 478. Spira MA, Lynch EC: Autoimmune hemolytic anemia and car-
thematosus. Q J Med 1991;80:605–612. cinoma: An unusual association. Am J Med 1979;67:753–758.
454. Sthoeger Z, Sthoeger D, Green L, Geltner D: The role of anti- 479. Bradley GW, Harvey M: Haemolytic anaemia with hyper-
cardiolipin autoantibodies in the pathogenesis of autoim- nephroma. Postgrad Med J 1981;57:46–47.
mune hemolytic anemia in systemic lupus erythematosus. J 480. Johnson P, Gualtieri RJ, Mohler DN, Carpenter JT:
Rheumatol 1993;20:2058–2061. Autoimmune hemolytic anemia associated with a hyper-
455. Xiros N, Binder T, Anger B, Bohlke J, Heimpel H: Idiopathic nephroma. South Med J 1985;78:1129–1131.
thrombocytopenic purpura and autoimmune hemolytic 481. Dacie JV: Auto-immune haemolytic anaemia (AIHA): Cold-
anemia in Hodgkin’s disease. Eur J Haematol 1988;40:437–441. antibody syndromes III: Haemolytic anaemia following
456. Eisner E, Ley AB, Mayer K: Coombs’-positive hemolytic mycoplasma pneumonia. In The Haemolytic Anaemias, 3rd
anemia in Hodgkin’s disease. Ann Intern Med 1967;66:258–273. ed. vol. 3, The Auto-Immune Haemolytic Anaemias. New
457. Levine AM, Thornton P, Forman SJ, et al: Positive Coombs test York: Churchill Livingstone, 1992:296–312.
in Hodgkin’s disease: Significance and implications. Blood 482. Perkin RL, Fox AD, Richards WL, King MH: Acute hemolytic
1980;55:607–611. anemia secondary to infectious mononucleosis. Can Med
458. Ertem M, Uysal Z, Yavuz G, Gozdasoglu S: Immune thrombo- Assoc J 1979;121:1095–1097.
cytopenia and hemolytic anemia as a presenting manifestation 483. Weikert LF, Davis RC: Profound anemia following a respira-
of Hodgkin disease. Pediatr Hematol Oncol 2000;17:181–185. tory infection. J Tenn Med Assoc 1995;88:270–271.
459. Shah SJ, Warrier RP, Ode DL, Lele HE, Yu LC: Immune throm- 484. Cherry JD: Anemia and mucocutaneous lesions due to
bocytopenia and hemolytic anemia associated with Hodgkin Mycoplasma pneumoniae infections. Clin Infect Dis
disease. J Pediatr Hematol Oncol 1996;18:227–229. 1993;17(Suppl. 1):S47–S51.
460. Kalmanti M, Polychronopoulou S: Autoimmune hemolytic 485. Bartholomew JR, Bell WR, Shirey RS: Cold agglutinin
anemia as an initial symptom in childhood Hodgkin’s disease. hemolytic anemia: Management with an environmental suit.
Pediatr Hematol Oncol 1992;9:393–395. Ann Intern Med 1987;106:243–244.
461. Weitberg AB, Harmon DC: Autoimmune neutropenia, 485a. Ness PM, Bell WR, Shirey RS: Transfusion medicine illus-
hemolytic anemia, and reticulocytopenia in Hodgkin’s trated. Novel management of cold agglutinin disease.
disease. Ann Intern Med 1984;100:702–703. Transfusion 2003;43:839.
462. Chu J-Y: Autoimmmune hemolytic anemia in childnood 486. Chu CS, Braun SR, Yarbro JW, Hayden MR: Corticosteroid
Hodgkin’s disease. Am J Pediatr Hematol Oncol 1982;4:125–128. treatment of hemolytic anemia associated with Mycoplasma
463. Economopoulos T, Stathakis N, Constantinidou M, pneumoniae pneumonia. South Med J 1990;83:1106–1108.
Papageorgiou E, Anastassiou C, Raptis S: Cold agglutinin 487. Turtzo DF, Ghatak PK: Acute hemolytic anemia with Myco-
disease in non-Hodgkin’s lymphoma. Eur J Haematol plasma pneumoniae pneumonia. JAMA 1976;236:1140–1141.
1995;55:69–71. 488. Lindstrom FD, Stahl-Furenhed B: Autoimmune haemolytic
464. Sivakumaran M, Murphy PT, Booker DJ, Wood JK, Stamps R, anaemia complicating Mycoplasma pneumoniae infection.
Sokol RJ: Paroxysmal cold haemoglobinuria caused by non- Scand J Infect Dis 1981;13:233–235.
Hodgkin’s lymphoma. Br J Haematol 1999;105:278–279. 489. Fiala M, Myhre BA, Chinh LT, Territo M, Edgington TS,
465. Sharara AI, Hillsley RE, Wax TD, Rosse WF: Paroxysmal cold Kattlove H: Pathogenesis of anemia associated with
hemoglobinuria associated with non-Hodgkin’s lymphoma. Mycoplasma pneumoniae. Acta Haematol 1974;51:297–301.
South Med J 1994;87:397–399. 490. Bowman HS, Marsh WL, Schumacher HR, Oyen R, Reihart J:
466. Domingo A, Crespo N, Fernandez dS, Domenech P, Jordan C, Auto anti-N immunohemolytic anemia in infectious mononu-
Callis M: Hairy cell leukemia and autoimmune hemolytic cleosis. Am J Clin Pathol 1974;61:465–472.
anemia. Leukemia 1992;6:606–607. 491. Green N, Goldenberg H: Acute hemolytic anemia and hemo-
467. Mainwaring CJ, Walewska R, Snowden J, et al: Fatal cold globinuria complicating infectious mononucleosis. Arch
anti-i autoimmune haemolytic anaemia complicating hairy Intern Med 1960;105:108.
cell leukaemia. Br J Haematol 2000;109:641–643. 492. Keyloun VE, Grace WJ: Acute hemolytic anemia complicat-
468. Cesana C, Brando B, Boiani E, et al: Effective treatment of ing infectious monocucleosis. N Y State J Med 1966;66:
autoimmune hemolytic anemia and hairy cell leukemia with 273–275.
interferon-alpha. Eur J Haematol 2002;68:120–121. 493. Tonkin AM, Mond HG, Alford FP, Hurley TH: Severe acute
469. Cohen Y, Polliack A, Zelig O, Goldfarb A: Monotherapy with haemolytic anaemia complicating infectious mononucleosis.
rituximab induces rapid remission of recurrent cold agglu- Med J Aust 1973;2:1048–1050.
tinin-mediated hemolytic anemia in a patient with indolent 494. Dacie JV, Auto-immune haemolytic anaemia (AIHA):
lympho-plasmacytic lymphoma. Leuk Lymphoma Cold-antibody syndromes IV: Haemolytic anaemia follow-
2001;42:1405–1408. ing infectious mononucleosis and other viral infections. In
470. Mori A, Tamaru J, Sumi H, Kondo H: Beneficial effects of The Haemolytic Anaemias, 3rd ed. vol. 3, The Auto-Immune
rituximab on primary cold agglutinin disease refractory to Haemolytic Anaemias. New York: Churchill Livingstone,
conventional therapy. Eur J Haematol 2002;68:243–246. 1992:313–328.
C H A P T E R 1 2

Immune Hemolysis
Associated with
Transplantation

Immune hemolysis caused by pendent of the human leukocyte antigen (HLA) gene
a variety of mechanisms has complex; consequently, even if the donor and recipi-
been noted after transplan- ent are HLA-matched, their RBCs are likely to be of
tation of hematopoietic stem different phenotypes.1-5 Of particular significance are
cells or solid organs.1,2 The those transplants between ABO blood group mis-
evaluation and appropriate matched donor-recipient pairs.
mangement of patients at risk
requires an understanding of Major and Minor Blood Group
both the manifold causes of
hemolysis and the settings in Incompatibility
which it can occur. ABO blood group incompatibility is defined as a
“major” incompatibility when the recipient’s immune
system is capable of producing antibodies against the
HEMATOPOIETIC CELL donor’s RBC antigens (e.g., group A donor and group
TRANSPLANTATION O recipient) and as a “minor” incompatibility when the
donor’s immune system is capable of producing anti-
The early transplantation literature regarding bodies against the recipient’s RBC antigens (e.g., group
hematopoietic cell transplantation referred only to O donor and group A recipient). In some instances,
bone marrow transplantation (BMT). More recently, both major and minor incompatibilities exist simulta-
there has been increasing use of peripheral blood stem neously—for example, when the donor is group B and
cells (PBSC) or stem cells obtained from umbilical the recipient is group A. Similar definitions can also be
cord blood. Although it is the patient’s bone marrow applied to other blood group systems.
that is reconstituted in each instance, we will maintain
the distinctions between BMT, PBSC transplants, and Autoantibodies and Alloantibodies
umbilical cord blood transplants where pertinent in
this chapter. The terms autoimmune and alloimmune must be used
Most cases of immune hemolysis associated with carefully with regard to antibodies that develop after
hematopoietic cell transplantation are related to the transplantation. An antibody produced by cells of the
fact that inheritance of blood group antigens is inde- donor against RBC antigens of the recipient should be

459
460 Immune Hemolytic Anemias

considered an alloantibody, as should antibodies


made by the recipient’s immune system against RBC TABLE 12-1. CLINICAL AND LABORATORY DATA
antigens of the donor. Donor antidonor or recipient REGARDING SIX PATIENTS WHO HAD BMT FROM
antirecipient antibodies, however, may appropriately A MINOR ABO-INCOMPATIBLE DONOR AND WHO
be defined as autoantibodies. DEVELOPED HEMOLYTIC ANEMIA IN THE
POSTTRANSPLANT PERIOD

MINOR ABO BLOOD ABO Group


GROUP–INCOMPATIBLE HEMATOPOIETIC Case and Rh Type Details of Hemolysis and
No. D R RBC Transfusion Requirement
STEM CELL TRANSPLANTS
1 0+ A– Severe intravascular hemolysis; hemoglobin-
uria; maximum hemolysis day +15;
The Passenger Lymphocyte Syndrome treated with plasma exchange; 9 units
RBCs required day +10 to +15
A well-recognized syndrome of immune hemolysis 2 0+ A+ Moderate hemolysis, maximum day +16;
after hematopoietic stem cell transplantation has 6 units RBCs required day +16 to +22
become known as passenger lymphocyte syndrome.1-3,6-17 3 0+ B+ Severe hemolysis; hemoglobinuria;
It occurs in some patients who are transplanted with maximum day +10; 14 units RBCs
hematopoietic stem cells from a minor ABO blood required day +10 to +19
4 0+ A+ Moderate hemolysis, maximum day +10;
group–mismatched donor. Occasionally, mismatches of 7 units RBCs required day +9 to +18
other blood groups result in similar (although milder) 5 B+ AB+ Moderate hemolysis; maximum day +12;
hemolysis. The syndrome has been attributed to prolif- 6 units RBCs required day +11 to +14
eration and antibody production by “passenger” lym- 6 A– A+ Moderate hemolysis, maximum day +13;
5 units RBCs required day +9 to +16
phocytes, which are infused with the stem cell product.
A typical case is illustrated in Figure 12-1, and clinical D-Donor; R-Recipient
and laboratory data regarding six patients described by From Petz LD: Immunohematologic problems unique to bone marrow
transplantation. In Garratty G (ed): Red Cell Antigens and Antibodies.
Hows and colleagues6 are listed in Table 12-1. (In one Arlington, VA: American Association of Blood Banks, 1986:195–229.
patient, the passenger lymphocyte syndrome was
cause by minor Rh incompatibility.)
Several factors can increase the risk of development 10; Haas and colleagues11 described a patient who
of the syndrome: the use of cyclosporine alone in the developed hemolysis on day 6; and, of the seven
absence of an antiproliferative agent for graft-vs.-host patients reported on by Gajewski and coworkers,12
disease (GVHD) prophylaxis; the use of peripheral hemolysis occurred between days 5 and 20 after
blood as a source of the hematopoietic stem cells; the transplantation.
use of a reduced-intensity pretransplant preparative Hemolysis is usually abrupt in onset and can be
regimen; utilization of a nongenotypically HLA- severe, with a rapidly dropping hemoglobin level,
matched donor and, possibly, use of a female donor.12,18 signs of intravascular hemolysis (hemoglobinemia and
Immune hemolysis generally has its onset near the hemoglobinuria),6,10-12,19 and renal failure.10,12,20-22 Less
end of the first week or during the second week after severe cases are characterized by a falling hemoglobin
transplantation. In the five patients described by level, increases in serum bilirubin and lactic dehydro-
Hows and coworkers,6 maximum hemolysis was genase (LDH), and decreased serum haptoglobin.
observed on days 10 to 16 after transplantation. Hemolysis usually persists for 5 to 10 days and
Rowley and Braine10 reported on five patients who then subsides as the patient’s residual incompatible
showed evidence of hemolysis between days 6 and RBCs are destroyed and are replaced by transfused

FIGURE 12-1. Minor ABO-mismatched BMT


RBCs Transfused 2 2 1 2 2 2 3 (donor group O, recipient group B). Flow
(0 + washed) chart shows hematologic and biochemical
14 HEMOGLOBINURIA evidence for hemolysis. Fourteen units of
packed group O RBCs were required to
12 1200 maintain the hemoglobin between day 10 and
day 19 post-BMT. Note that the hemoglobin,
LDH (IU/L)

Total Bilirubin (mg/dL)

10 1000 10 the lactic dehydrogenase, and the total


8 800 8 bilirubin were normal for the first 5 days
Hb (g/dL)

post-BMT. During the hemolytic episode, no


6 600 6 incompatible isohemagglutinins were infused;
RBCs transfused were washed group O,
4 400 4
platelets were group B or washed and
2 200 2 resuspended in fresh AB plasma. (From
Hows J, Beddow K, Gordon Smith E, et al:
Donor-derived red blood cell antibodies and
0 5 10 15 20 25 30
immune hemolysis after allogeneic bone
Days post-BMT marrow transplantation. Blood
1986;67:177–181.)
Immune Hemolysis Associated with Transplantation 461

group O RBCs and/or by RBCs of donor type pro- Apparently, significant hemolysis can occur at a
duced by cells derived from the engrafted stem cells. time when the amount of anti-A or anti-B is still too
Also, antibody production gradually subsides as the small to be detectable by routine serologic tests (also
passenger lymphocytes that are not engrafted reach see pages 469–470). Similar findings have been reported
the end of their life span. after solid organ transplantation.24
Measuring anti-A and anti-B titers in the donor
before transplantation apparently does not help to
SEROLOGIC FINDINGS predict either which patients will develop passenger
Table 12-2 displays the results of serological tests for lymphocyte syndrome or the severity of hemolysis.6
the six patients described by Hows and associates.6 Passenger lymphocyte syndrome is most likely to
The direct antibody test (DAT) first became positive on occur when the donor is group O and the patient
days 4 to 18 after transplanation, and anti-A and/or group A, a phenomenon which perhaps is related to
anti-B were found in eluates from the patients’ RBCs the fact that IgG anti-A and anti-B are found far more
and in the patients’ sera. In four patients, the DAT was commonly in group O than in group B or A subjects.25
positive with anti-C3 but negative with anti-IgG. It is also true that anti-A–sensitized RBCs bind C1q
Nevertheless, a red cell eluate made from RBCs of two (the earliest reacting component of complement)
patients with this DAT pattern revealed anti-A. three to six times more than do anti-B–sensitized
Of particular interest is that maximum hemolysis RBCs26 and that an equal level of hemolysis is
occurred in one patient on the 15th day after trans- obtained in vitro only if the fluid phase anti-B is
plantation, but the DAT did not become positive until present in concentrations two to four times higher
day 18. Our subsequent experience has confirmed than that of anti-A.27
such findings in that signs of hemolysis might precede
one’s ability to detect the expected antibody by 1 or ANTIBODIES OTHER THAN THOSE OF THE ABO
2 days. Similar observations have been reported by BLOOD GROUP SYSTEM PRODUCED BY
Greeno and colleagues,20 who reported on a patient PASSENGER LYMPHOCYTES
who developed marked hemolysis on the eighth day
after transplanation, although serum anti-A was not Hows and colleagues6 evaluated seven D-positive
detectable until the 10th day. Leo and coworkers23 patients receiving marrow from D-negative donors
emphasized that in their patient, the clinical signs of and found that three developed donor-derived anti-
hemolysis—including a decrease in hemoglobin and a D and one of these also developed donor-derived
rise in LDH—preceded the detection of the relevant anti-C and anti-E. Only one of the patients had hemo-
antibody. lysis. This patient had severe aplastic anemia, was
Also, Tiplady and associates16 described a patient group A, Rh positive, and was transplanted with
who developed signs of hemolysis with a drop in marrow from a donor who was group A, Rh-negative
hemoglobin from 10.3 g/dL to 8.3 g/dL on the eighth and whose serum contained anti-D. No antibody was
day after a minor ABO-incompatible peripheral blood detected in the patient’s serum until the 13th day after
stem cell transplant, although no antibody was transplantation, at which time anti-D was found.
detectable at that point and the DAT was still nega- Moderate hemolysis developed, which reached its
tive. On day 9, the hemoglobin fell further to 6.3 g/dL, maximum on day 13, and the patient required trans-
and although the DAT was now positive, anti-A was fusion of 5 units of RBC on days 9 to 16. The antibody
not detectable until the next day. titer rose to 256 on day 70; later samples were not

TABLE 12-2. SEROLOGICAL INVESTIGATION OF PATIENTS PRESENTING WITH IMMUNE HEMOLYSIS

Maximum Day
Hemolysis ABO Group DAT Post-BMT Day Post-BMT
Case Day Rh Type Positive DAT Antibody Antibody Serum Antibody
No. Post-BMT D R for First Positive in Eluate in Serum Last Detected

1 +15 O+ A– C3 +18 ND Anti-A +38*


2 +16 O+ A+ C3 +15 ND Anti-A +24*
3 +10 O+ B+ IgG + C3 +10 Anti-B Anti-B +42*
4 +10 O+ A+ C3 +4 Anti-A Anti-A +30*
5 +12 B+ AB+ C3 +9 Anti-A Anti-A +42*
6 +13 A– A+ IgG +13 Anti-D Anti-D +70†

DAT, direct antiglobulin test; D, donor; R, recipient; ND, not done.


* Serum antibody not detectable in subsequent follow-up.
† Anti-D titer 256; later samples not tested.
From Hows J, Beddow K, Gordon Smith E, et al: Donor-derived red blood cell antibodies and immune hemolysis after allogeneic bone marrow transplantation. Blood
1986;67:177–181.
462 Immune Hemolytic Anemias

tested. The donor-derived anti-D produced in this were anti-D, -E, and -s. The authors pointed out that
recipient was, therefore, the result of a secondary cases of non-ABO immune hemolysis frequently
immune response to the D antigen by the donor’s pas- involve donors and recipients who have no prior evi-
senger lymphocytes. dence of alloantibodies, thus making it impossible to
No evidence for donor presensitization to Rh anti- predict those patients who are at risk.
gens was present, however, in one of the cases Franchini and colleagues31 described a patient who
reported by Hows and coworkers,6 in which donor- developed anti-D of donor origin and who showed
derived Rh antibodies were produced. This donor signs of mild hemolytic anemia after a BMT.
was a 15-year-old boy who had not been transfused,
suggesting that in this case the production of donor-
LEWIS BLOOD GROUP SYSTEM
derived anti-D, -C, and -E after transplantation was
due to a primary immune response by the donor lym- A number of authors have pointed out that the
phocytes. The donor for the third patient, who devel- Lewis antigens always remain of the recipient type
oped anti-D post-transplant, had no Rh antibodies after BMT.32-35 This finding is consistent with the fact
detectable in her serum, but she had two children who that Lewis antigens are absorbed by RBCs from the
were D positive. plasma and are not synthesized by RBCs or their
Robertson and associates28 found anti-Jkb in the precursors.36
serum and in an eluate of a Jk (b+) recipient 9 days Myser and coworkers32 described a patient who
after transplantation of a T-cell depleted marrow from developed hemolytic anti-Lea 8 days after BMT. The
a Jk (b–) donor. No anti-Jkb was found in the donor patient was Le (a-b+) and, at the time of the BMT, a
before BMT, but she might have been sensitized hemolytic anti-Lea was detected in the Le (a-b-) donor.
through pregnancy. The patient did not develop signs No hemolysis resulted, but the anti-Lea interfered with
of hemolysis. HLA antibody identification in that strong lymphocyto-
Ting and colleagues29 reported production of non- toxic reactions occurred with Le (a+) donors.
ABO RBC alloantibodies in 12 of 150 patients after
BMT. Although none of the antibodies caused hemoly-
sis, the researchers’ report is particularly interesting in SOURCE OF ANTIBODY CAUSING HEMOLYSIS
that it emphasizes the different sources of the anti-
Detailed serologic studies indicate that the relevant
bodies (also see pages 488–490). Passenger lympho-
serum antibody is not present in the period immedi-
cytes were the evident source of antibodies in two
ately after transplantation but rather is first detectable
patients who developed antibodies within 19 days of
at about the time hemolysis becomes evident.1-3,5,6
transplantation.
Therefore, passive transfer during infusion of the
Leo and coworkers23 reported severe hemolysis
donor’s plasma with the stem cell product cannot
caused by anti-Jka after a PBSC transplant from an
account for the presence of the antibody. Also, anti-
HLA-identical sibling donor. The case is of particular
body production and hemolysis generally occur
interest, as prophylaxis against GVHD consisted of
before clinical evidence of engraftment while pancy-
methotrexate (administered on days 5 and 7 after trans-
topenia caused by the pretransplant preparative
plantation at a dose of 10 mg/m2), and cyclosporine
regimen is present and before immune reconstitution
(begun on the day before transplantation at a dose of
of the patient. Lymphocytes derived from engrafted
5 mg/kg). A myeloablative conditioning regimen was
donor lymphoid cells probably would not be present
used, which included total body irradiation. Hemolysis
ensued on the 18th day after transplantation, and the so soon after transplantation.8 Instead, the syndrome
has been attributed to the production of antibody by
patient was managed with RBC transfusions. The
rapidly proliferating “passenger” lymphocytes trans-
authors pointed out that their case illustrated that clin-
fused with the donor stem cell product, which are
ical signs of hemolysis, such as a decrease in hemoglo-
present temporarily until the end of their life spans.
bin or a rise in LDH, can precede the serologic detection
Pertinent data regarding the source of antibody pro-
of the responsible antibody.
duction has come from study of passenger lympho-
Young and associates30 also reported on two patients
cyte syndrome after solid organ transplantation.
who developed the passenger lymphocyte syndrome
Analysis of the immunoglobulin allotypes of the rele-
caused by anti-Jka. Hemolysis was abrupt in onset and
vant antibodies and comparison with the allotypes of
severe, with rapidly dropping hemoglobin, signs of
the donor and recipient have documented that the
intravascular and extravascular hemolysis, and renal
antibodies are produced by antibody-producing cells
failure. One of the transplants used stem cells from the
of donor origin (see page 500).
bone marrow of an unrelated male HLA-matched
donor, and the other case was a PBSC transplant from
an HLA-matched male sibling. Both transplants used a FREQUENCY OF DONOR-DERIVED ANTIBODY
myeloablative conditioning regimen. They also identi- PRODUCTION AND HEMOLYSIS
fied three additional cases in a review of hospital
records involving 427 allogeneic stem cell transplants A retrospective analysis of 21 consecutive cyclosporine-
over a 6-year period; antibodies in these three cases treated BMT patients receiving marrow lacking A, B, or
Immune Hemolysis Associated with Transplantation 463

D antigens that were present in the recipient showed • An eluate revealed anti-A or anti-B in three of the
that 15 of 18 patients tested had red cell antibody pro- four instances in which this was tested.
duction against recipient red cell antigens. Despite the
frequent presence of donor-derived antibody, however, Three of the patients had massive hemolysis. The
only 3 of 21 patients (12.5%) developed clinically course of one of these three patients is illustrated in
significant hemolysis.6 None of the marrow donors had Figure 12-2.
a high titer of anti-A or anti-B, with the highest IgG/
IgM titers being 64/128. Transfusion Requirements Exceeding
Hemolytic anemia was more frequent in a series
reported by Rowley and Braine,10 who found that the Recipient’s RBC Volume
hemolysis developed in five of seven evaluable minor Transfusion requirements for the three patients with
ABO-mismatched transplants. Hemolysis was severe massive hemolysis were far greater than could
in several patients—two developing renal failure and be accounted for by lysis of the patients’ ABO-
one requiring hemodialysis. incompatible RBCs12 (Table 12-3). The patients’ RBC
Hazlehurst and associates7 reported that two of volumes on the fifth day after transplantation
nine recipients (22%) of T-cell–depleted minor ABO- ranged from 1592 mL to 2039 mL, whereas the
incompatible marrow grafts developed hemolysis. volume of RBCs transfused for each patient was
Robertson and colleagues13 reported that two of ten 4680 mL (26 units of RBCs at 180 mL/unit). All of the
group A patients (20%) receiving T-cell–depleted units of transfused RBCs were group O and were
marrows developed hemolysis, whereas none of five saline washed.
group B patients receiving T-cell–depleted marrows The authors also determined that when the donor
and none of six patients receiving marrows without and recipient were ABO compatible, the “baseline”
the removal of T cells (three group A, one group AB, RBC transfusion requirement for days 5 to 20 in
and two group B) developed ABO blood group anti- marrow transplant patients at the same medical center
bodies or hemolysis. Bensinger and colleagues37 averaged 0.85 times the patient’s RBC volume on day 5.
reported only two episodes of acute hemolysis among This transfusion requirement results from marrow
87 patients transplanted with ABO-incompatible hypoplasia caused by ablative pretransplant
marrow. radiochemotherapy; in addition, there could be some
Young and coworkers30 identified a total of five occult bleeding caused by the hypoproliferative throm-
patients with hemolysis involving non-ABO red cell bocytopenia. It is also possible that RBC survival is not
alloantibodies among a total of 427 allogeneic normal after such high-dose radiochemotherapy,
hematopoietic stem cell transplants, for an incidence although this has not been measured.
of approximately 1%. The transfusion requirements for group O RBCs in
the three patients with massive hemolysis exceeded the
PATIENTS WITH MASSIVE HEMOLYSIS patients’ volume of ABO-incompatible RBCs plus the
“baseline” transfusion requirement by 908–1735 mL
Of particular concern are the occasional reports of (mean = 1433 mL) (see Table 12-3). Accordingly, even if
massive hemolysis after minor ABO-incompatible all of the patients’ ABO-incompatible RBCs had been
marrow or peripheral blood stem cell allotransplanta- hemolyzed and consideration had been given to the
tion12,14,16,19-22 that lead to renal insufficiency and even baseline transfusion requirement (as when there is no
fatal multisystem organ failure.12,20,21,38 ABO incompatibility), an average of almost 1500 mL of
Gajewski and associates12 described striking hemo- additional group O RBCs needed to be transfused. As
lysis in seven patients transplanted with minor ABO- none of the patients demonstrated any signs of bleed-
incompatible marrow grafts from matched unrelated ing, the inescapable conclusion was that rather large
donors. In four of the patients, the marked hemolysis volumes of transfused group O RBCs were hemolyzed
was accompanied by renal failure severe enough to in addition to the patients’ own RBCs.
require hemodialysis. The episodes of hemolysis were
typical of the passenger lymphocyte syndrome: Bystander Hemolysis
• Hemolysis began 5 to 8 days after transplantation Because the only RBC antibodies detected in these
and was manifested by a decreasing hematocrit, patients were anti-A and anti-B and yet group O RBCs
elevated bilirubin, elevated LDH, and an were also hemolyzed, it appears that hemolysis of
increased transfusion requirement. antigen-negative RBCs occurred. We refer to this phe-
• Hemolysis persisted for 2 to 12 days. nomenon as bystander immune hemolysis, which we
• A strongly reactive (3+ or 4+) anti-A or anti-B, define as immune hemolysis of cells that are intrinsi-
consistent with donor lymphocyte origin, was cally negative for the antigen against which the rele-
found in the serum of each patient. vant antibody is directed.5,12
• No other allo- or autoantibodies were present. Transplant physicians have been cautious regarding
• The DAT was positive during hemolysis in all their acceptance of the concept of bystander immune
cases. hemolysis, but there has been an increasing number of
464 Immune Hemolytic Anemias

Patient R.L (Recipient Group A/Donor Group O)


RBC 40
Tsf
35
30
HCT 25
%
20
15

8
7
6
T. Bili 5
mg/dL 4
3
2
1
0
PS 0 +m +m +m 0
DAT lgG NT 0 0 0 NT
C3 NT 0 0 0 NT
Anti-A 0 0 2+ 2+ 4+ 4+ 4+ 3+ 4+

–1 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25
FIGURE 12-2. Flow chart of index group patient not receiving pretransplant exchange transfusion showing
transfusion requirements with biochemical and serologic evidence for hemolysis. RBC Tsf, RBC
transfusions (arrows); DAT, direct antiglobulin reagent (anti-IgG + C3); PS, polyspecific; IgG, anti-IgG reagent;
4+, cells agglutinated in one clump; 3+, cells agglutinated in three to six very large clumps; 2+, cells
agglutinated in many medium clumps; 1+, cells agglutinated in numerous tiny clumps, very cloudy
background; +m, very tiny agglutinates only seen microscopically; 0, no agglutinates detected; NT, not
tested; ↓, transfusion. (From Gajewski JL, Petz LD, Calhoun L, et al: Hemolysis of transfused group O red
blood cells in minor ABO-incompatible unrelated-donor bone marrow transplants in patients receiving
cyclosporine without posttransplant methotrexate. Blood 1992;79:3076–3085.)

reports of massive hemolysis that is far more exten-


TABLE 12-3. TRANSFUSION REQUIREMENTS sive than can be explained on the basis of hemolysis of
OF GROUP O RBCs IN THREE PATIENTS the patient’s own RBCs. Accordingly, bystander
TRANSPLANTED WITH BONE MARROW FROM immune hemolysis has been discussed more fre-
UNRELATED MINOR ABO-INCOMPATIBLE DONORS quently in more recent medical literature.15-17,39
Bystander immune hemolysis may also occur in a
Vol. of Baseline Excess
Patient’s RBCs Transfusion Transfusion
number of other clinical settings, and there are several
RBC Transfused Requirement* Requirement† possible mechanisms.5 See Chapter 9 for a detailed
Volume Day Day +5 to Day +5 to Day +5 to discussion of this topic and Chapter 14 for a discus-
Patient +5 (mL) +20 (mL) +20 (mL) +20 (mL) sion of its possible role in the sickle cell hemolytic
transfusion reaction syndrome.
1 2039 4680 1733 908
2 1592 4680 1353 1735
3 1635 4680 1389 1656 Additional Reports of Severe Hemolysis
* Baseline transfusion requirement was determined by measuring the transfusion Greeno and coworkers20 reported on a patient who
requirements during days +5 to +20 in 61 marrow transplant patients in whom
the donor and recipient were ABO identical. This value was 0.85 times the hemolyzed her entire group A RBC population
RBC volume on day +5.
† Excess transfusion requirement is defined as the volume of group O RBCs
between the 8th and 11th days after transplantation.
required above the sum of the baseline transfusion requirement and the patient’s
By day 11, the circulating RBCs were completely
volume of (ABO incompatible) RBCs on day +5. Thus, even if all of the group O, but she continued to require transfusion of
patient’s ABO-incompatible RBCs were hemolyzed and there is, in addition, a 2 units of group O RBCs every 2 to 3 days for an
baseline transfusion requirement, an average of almost 1.5 liters additional
group O RBCs needed to be transfused. Therefore, not only were the patient’s additional 10 days, for a total of 16 units of RBCs
ABO-incompatible RBCs hemolyzed, but transfused group O RBCs must have (about 2800 mL of packed RBCs) (Fig. 12-3). The
been hemolyzed as well. patient’s calculated group A RBC volume on day 7
From Petz LD: The expanding boundaries of transfusion medicine. In Nance SJ
(ed): Clinical and Basic Science Aspects of Immunohematology. Arlington, VA: was only about 1000 mL. The authors pointed out
American Association of Blood Banks, 1991:73–113. that the mechanism of the hemolysis of the trans-
fused group O RBCs is unknown.
Immune Hemolysis Associated with Transplantation 465

50 50 Worel and associates39 described four patients who


Transplant developed passenger lymphocyte syndrome, one of
whom received 40 units of RBC during the first 30
40 40
days after transplantation of a minor ABO-incompati-
ble transplant using peripheral blood stem cells and a
30 30 nonmyeloablative conditioning regimen. The patient

Bilirubin (mg/dL)
ultimately died of multiorgan failure on day 35. The
Percentage

authors commented that the extent of hemolysis sug-


20 20 gested bystander hemolysis in addition to immune
hemolysis due to ABO incompatibility.
Herzog and coworkers40 reported a patient of group
10 10 A1 who was transplanted with marrow from a group
A2 donor. On the eighth day after transplantation,
coincident with transfusion of 2 units of group A1
0 0
RBC, the patient developed intravascular hemolysis
DAT: N N NPNN N N N
and massive hemoglobinuria. During the next 2
–10
weeks, transfusion of 14 units of group O RBCs was
–8 –4 0 4 8 12 16 20 required. The only antibody detectable was anti-A1.
Transplant day The authors did not comment on the fact that the
FIGURE 12-3. Shown is the course of the patient’s hematocrit (- 䊐 -)
volume of transfused group O RBCs (about 4000 mL)
and total bilirubin (-o-) during her second transplant. The type A must have exceeded the volume of the patient’s
hematocrit (--- 䊐 ---) was calculated by multiplying the percentage of A incompatible RBCs (including the transfused group
RBCs by the total hematocrit. Each solid arrow represents the A1 RBC) that were present at the onset of hemolysis.
transfusion of one unit of RBCs. Along the bottom of the graph are Bolan and colleagues18 reported massive immune
shown the results of the DAT, with N representing negative and P
representing positive. (From Greeno EW, Perry EH, Ilstrup SJ, Weisdorf hemolysis in three out of ten consecutive patients
DJ: Exchange transfusion the hard way: Massive hemolysis following undergoing HLA-identical, related-donor PBSC
transplantation of bone marrow with minor ABO incompatibility. transplants with minor ABO incompatibility.
Transfusion 1996;36:71–74.) Nonablative conditioning had been given to nine of
these ten patients, including two patients with
hemolysis. Cyclosporine alone was used as prophy-
laxis against GVHD. Catastrophic hemolysis of 78%
Tiplady and colleagues16 described a 28-year-old of the circulating red cell mass led to anoxic death in
man with lymphoblastic lymphoma who received the first patient they observed (Fig. 12-4). In their
G-CSF mobilized stem cells from his HLA-identical second patient, the authors calculated that hemolysis
sister. The recipient’s blood group was A Rh D of more than 80% of the patient’s estimated red cell
positive, and the donor’s group was O Rh D positive. mass had occurred during a 36-hour period. In their
The patient received cyclosporine (but not methotrex- third patient, the volume of red cells transfused
ate) for GVHD prophylaxis. On the eighth day after between days 5 and 11 was equivalent to the patient’s
transplantation, his hemoglobin fell from 10.3 g/dL to entire estimated red cell volume. The patient also
8.3 g/dL, but the DAT was negative and no anti-A was required “numerous” additional RBC transfusions,
detectable. Three units of group A blood were trans- the need for which was attributed to the development
fused without complications. On day 9, the hemoglo- of pulmonary hemorrhage on day 13 and thrombotic
bin fell further to 6.3 g/dL and his DAT became microangiopathy on day 26. The authors did not
positive (IgG and C3d), but anti-A was not detectable seem to accept the notion that transfused group O
until day 10. From day 9 to day 13, he received 17 units RBCs were being hemolyzed.
of group O blood (approximately 3 L of RBCs) at a time Haas and associates11 reported on a group A1
when his calculated RBC volume was 1 L. By day 26, a patient who was transplanted with a T-cell–depleted
further 8 units of blood had been transfused, bringing bone marrow from a group O donor. The patient
the total to 28. He had no appreciable blood loss, his developed a positive DAT, and anti-A1 was found in
bilirubin level remained elevated, and his response to the serum. On the sixth day after transplantation, the
transfusion was short lived. His blood film did not patient developed life-threatening intravasclar hemo-
show any evidence of microangiopathic hemolysis. lysis, with the hemoglobin dropping from 11.8 g/dL
The excessive transfusion requirements led the authors to 3.8 g/dL in 6 hours (Fig. 12-5).
to speculate that the hemolysis of compatible blood There have been a number of cases of severe hemo-
was occurring, as only anti-A and anti-B were ever lysis after PBSC transplantation in addition to those
found in the patient’s serum. They commented that reported by Bolan and coworkers18 and described
bystander hemolysis seems to be a very rare, albeit previously. Salmon and colleagues41 reported a 16-year-
clinically serious complication of hematopoietic stem old boy who underwent an allogeneic PBSC transplant
cell transplantation. from his mother who was of a different ABO group
466 Immune Hemolytic Anemias

35 14000

30 12000
FIGURE 12-4. Course of severe hemolysis in a patient. On
days 8 and 9 after transplantation, fever, hemodynamic
25 10000
HCT (%), WBC (10g/L)

instability, and renal insufficiency occurred, the hematocrit


HCT dropped preciptously, and the LDH doubled. Although the

LHD (U/L)
20 8000 hematocrit responded to transfusions with group O red cells,
WBC
cardiopulmonary arrest occurred, renal failure ensued,
LDH neurological function did not recover, and the patient died on
15 6000
RBC Transfusions day 16. Serological testing revealed that donor-type anti-A
isohemagglutinins were the cause of massive immune
10 4000
hemolysis. (From Bolan CD, Childs RW, Procter JL, Barrett
AJ, Leitman SF: Massive immune haemolysis after allogeneic
5 2000 peripheral blood stem cell transplantation with minor ABO
incompatibility. Br J Haematol 2001;112:787–795.)
0 0
5 6 7 8 9 10 11
Days after PBSC infusion

(mother group O, Rh+, patient group A, Rh+). Hemo- using anti-C3d, and IgM and IgG anti-A antibodies
lysis developed abruptly on day 9, and all group A red were found in the patient’s serum with a titer of 8000.
cells of the recipient were destroyed within 36 hours. He recovered after being treated with an exchange
Oziel-Taieb and associates21 described a group A transfusion of one blood volume, 6 units of packed red
patient who received a PBSC transplant from a group cells, fluids, and diuretics.
O donor and developed immune hemolysis starting Laurencet and colleagues14 described “massive”
on day 9. Anti-A was found in the patient’s serum and hemolysis in a 37-year-old man beginning on day 12
in an eluate from his RBCs. After the onset of hemoly- after a PBSC transplant with minor ABO incompatibil-
sis, the patient was transfused with 10 units of group ity. Hemolysis was marked, as indicated by the fact that
O RBCs over the next 10 days, but he developed acute his hematocrit dropped from 30.1% on day 0 to 18.8%
renal failure and acute respiratory distress syndrome on day 12. The patient required transfusion of only
and died on day 20 from multiorgan failure. 6 units of group O RBCs, however, and he recovered.
Toren and coworkers19 described a 12-year-old boy, The patient reported on by Bornhauser and associ-
blood group A, who received an allogeneic PBSC trans- ates42 received a G-CSF–mobilized PBSC from an un-
plant from his HLA-matched sister, blood group O. On related HLA-matched donor. The blood groups of the
day 8, he developed fever, abdominal and bilateral patient and donor were bidirectionally incompatible.
flank pain, and severe intravascular hemolysis with Signs of “major hemolysis” developed on the fifth day
hemoglobinemia and hemoglobinuria and a decrease after transplantation, and the patient’s hemoglobin
in his hemoglobin to 5.4 g/dL. The DAT was positive reached a nadir of 4.3 g/dL.

CMV
1st BMT 2nd BMT
H. GL.
14 RBC A1 A2 0 0 0
washed

FIGURE 12-5. Hemoglobin (Hb); ×—×;


10 reticulocytes: ×---×; granulocytes: •—•,
GRANULOCYTES. of the transplant patient. BMT = bone
marrow transplantation; CMV H.GI. =
/mm3
Hb g/dI

1200 0.4 cytomegalovirus hyperimmune globulin;


RBC = red blood cells. (From Haas RJ,
Rieber P, Helmig M, Strobel E,
% RETIC.

6 800 0.3 Belohradsky BH, Heim MU: Acquired


immune haemolysis by anti-A1 antibody
600 0.2 following bone marrow transplantation.
Blut 1986;53:401–404.)

2 400 0.1

29 31 2 4 6 8 10 12 14 16 18 20
October 1985 November 1985
Immune Hemolysis Associated with Transplantation 467

Delamaire and coworkers43 described a patient who lative peripheral blood stem cell transplants (23
had fatal hemolysis after PBSC transplantation. patients with related donors and 17 with unrelated
donors). They commented that the high incidence
reported by Bolan and associates18 could be due to
THE SOURCE OF HEMATOPOIETIC STEM CELLS IN differences in the kind of postgrafting immuno-
PATIENTS DEVELOPING PASSENGER suppression used, as Bolan and associates used
LYMPHOCYTE SYNDROME cyclosporine only, whereas Mielcrek and coworkers
used cyclosporine in combination with the anti-
It is possible that the passenger lymphocyte syn- metabolite mycophenylate mofetil. The authors
drome is more common and more severe among commented that the passenger lymphocyte syn-
patients who have received PBSC transplants com- drome might occur in a minority of mycophenylate
pared with those who have received marrow trans- mofetil/cyclosporine-treated nonmyeloablative reci-
plants.14,15,18,19,21,22,39,41-43 As will be reviewed below pients, whereas it is quite rare among patients
regarding the passenger lymphocyte syndrome occur- treated with methotrexate.
ring after solid organ transplants, the incidence and Worel and colleagues39 reported on four patients
severity of hemolysis in minor ABO-mismatched trans- who developed severe immune hemolysis after minor
plants are related directly to the lymphoid content of ABO-incompatible allogeneic PBSC transplantation.
the grafted organ.18,44,45 PBSCs contain a 16-fold Three of the patients had undergone a nonmyeloabla-
increase in CD3+ T lymphocytes and an 11-fold increase tive conditioning regimen. Hemolysis occurred in one
in CD19+ B lymphocytes compared with conventional of 15 patients (6%) after myeloablative conditioning
marrow harvests.46 Also, lymphocytes in PBSC grafts and in three of ten patients (30%) after dose-reduced
might be specifically primed for antibody production, conditioning, and only in patients not receiving
owing to a switch from a type 1 to a type 2 helper methotrexate for GVHD prophylaxis. The authors sug-
response induced by the administration of granulocyte gested that patients receiving minor or bidirectional
colony-stimulating factor.47 ABO-incompatible PBSC grafts in combination with
Lapierre and colleagues48 studied the significance of GVHD prophylaxis regimens without methotrexate
the hematopoietic stem cell source and concluded that are at risk of severe hemolysis.
immunohematologic reconstitution differed signifi- Reduced-intensity pretransplant regimens fre-
cantly after granulocyte colony-stimulating factor- quently use fludarabine, which could be an etiologic
mobilized PBSC transplantation compared with bone factor in the development of RBC autoantibodies
marrow stem cell transplantation. They suggested that among patients receiving the drug (see Chapter 8).
such a difference could contribute to acute hemolysis The role of fludarabine in the development of
described after PBSC transplantation. antibodies in passenger lymphocyte syndrome is
As of this writing, no instances of passenger lym- uncertain.
phocyte syndrome have been reported after umbilical T-Cell Depletion. Hazlehurst and associates7
cord blood hematopoietic cell transplants. reported the development of anti-A/B in eight of nine
patients transplanted with T-cell–depleted marrow,
none of whom had received cyclosporine. The DAT
SIGNIfiCANCE OF IMMUNE MODULATION was positive for five patients, and two of these had a
AS A CAUSE OF PASSENGER fall in hemoglobin of up to 2 g/dL. Also, Haas and
LYMPHOCYTE SYNDROME coworkers11 reported one patient and Robertson and
colleagues13 reported two patients who were group A1,
The Pretransplant Conditioning Regimen. Two of
received a T-cell–depleted marrow from a group O
the three patients reported on by Bolan and associates18
donor but did not receive cyclosporine, and developed
had received low-intensity nonmyeloablative pre-
significant hemolysis due to anti-A or anti-A1.
transplant conditioning regimens. Such regimens are
being used increasingly in hematopoietic stem cell
transplantation. Often, cyclosporine is used as the sole
agent for GVHD prophylaxis, and methotrexate or
other antiproliferative agents are deliberately avoided
Post-transplant Graft-Vs.-Host-
with the goal of enhancing immune-mediated donor Disease Prophylaxis
antitumor effects. Because the conditioning regimen CYCLOSPORINE/METHOTREXATE
and the anti-GVHD regimen heavily influence engraft-
ment, GVHD, tumor eradication, and the prevention of Cyclosporine is a powerful immunosuppressive agent
relapse, altering these regimens for the purposes of that suppresses lymphocyte proliferative responses to
prevention of passenger lymphocyte syndrome is not mitogens and alloantigens in a dose-dependent
recommended.18 fashion. Primary responses are much more sensitive to
Mielcarek and coworkers 49 reported that the the effects of cyclosporine than are secondary
passenger lymphocyte syndrome occurred in only responses.50 T-lymphocyte proliferation is not impaired
two of 40 patients with HLA-matched nonmyeloab- when presensitized lymphocytes are re-exposed to
468 Immune Hemolytic Anemias

antigen in the presence of cyclosporine.6 Although she underwent a second BMT using marrow from the
most patients who have developed hemolysis as a same donor. Continuous tacrolimus infusion begin-
result of passenger lymphocyte syndrome have ning one day before the marrow transplant was the
received cyclosporine for immunosuppression, its only GVHD prophylaxis given. As described pre-
precise role in facilitating donor-derived antibody pro- viously, the patient hemolyzed her entire red cell
duction is uncertain.6 mass between days 8 and 11 after transplantation as a
In all six cases of the passenger lymphocyte syn- result of anti-A produced by passenger lymphocytes.
drome reported by Hows and associates,6 the patients The authors suggested that tacrolimus could be asso-
were treated with cyclosporine for GVHD prophy- ciated with only modest immunosuppressive activity
laxis after BMT. In contrast, a retrospective review of against donor-derived passenger lymphocytes, but
13 patients in whom methotrexate was used instead of that it might effectively suppress T-cell regulation of
cyclosporine revealed no cases of immune hemolysis B-cell proliferation.
due to donor-derived antibody. Thus, available data indicate that donor-derived
As described by Gajewski and coworkers,12 marked antibody production and hemolysis could occur in
hemolysis occurred among a series of patients receiv- association with immune modulation caused by
ing minor ABO-incompatible marrow grafts that were cyclosporine, tacrolimus, or T-cell depletion of the
not T-cell depleted and that were from unrelated marrow. Additional risk factors might be the use of
donors. Seven consecutive patients who received peripheral blood stem cells, a nonmyeloablative con-
received cyclosporine without methotrexate for post- ditioning regimen, use of nongenotypically HLA-
transplant GVHD prophylaxis developed the passen- matched donors, and the use of female donors.18
ger lymphocyte syndrome with severe hemolysis. In
contrast, no hemolysis was observed among similar
patients who received both cyclosporine and metho- MANAGEMENT OF PATIENTS
trexate, although weakly positive tests for anti-A or RECEIVING HEMATOPOIETIC CELL
anti-B were found in two of seven patients so treated. TRANSPLANTS FROM MINOR
The authors suggested that post-transplant immuno-
suppression with methotrexate might prevent the pas- ABO-INCOMPATIBLE DONORS
senger lymphocyte syndrome from occurring even
among patients receiving cyclosporine. Indeed, Volume Reduction of Marrow Products to
methotrexate has been shown to be cytotoxic for B Prevent Hemolytic Transfusion Reaction
lymphocytes and might prevent the antibody produc-
tion necessary for the passenger lymphocyte syndrome Because the volume of a bone marrow product for a
to occur.51,52 Rowley15 indicated that special attention transplant recipient can be 700 mL or greater, one
must be devoted to the recipient if antiproliferative must be concerned that the isohemagglutinins in the
agents for GVHD prophylaxis (e.g., methotrexate, donor plasma might cause an acute hemolytic trans-
trimetrexate, and mycophenylate mofetil) are not used fusion reaction. One option is to measure the anti-A
as part of the GVHD prophylactic regimen. and/or anti-B titer of each marrow donor when there
With the exception of the patient reported on by is a minor ABO incompatibility with the intended
Bornhauser and colleagues,42 cyclosporine without recipient. If the titer of the relevant ABO antibody is
methotrexate was used for post-transplant GVHD greater than 256, one should consider the advisability
prophylaxis for all of the patients described previ- of plasma reduction of the marrow product. The deci-
ously who experienced severe hemolysis after PBSC sion is reached on the basis of the height of the iso-
transplants. hemagglutinin titer, the adequacy of the marrow
Donor-derived antibody, however, has been harvest, and the knowledge that some stem cells will
detected in a number of patients when other means of be lost during manipulation of the marrow.
immune modulation have been used. For example, Lasky and colleagues54 reported that plasma reduc-
Bar and associates53 found donor-derived antibody tion of donor marrow products in five cases resulted
without hemolysis in three recipients of minor in a reduction of the original marrow volume by 54%
ABO-incompatible marrow transplant patients who and a reduction in plasma volume by 71%. The trans-
received cyclophosphamide rather than cyclosporine fused marrow contained a mean of 99.5% of the origi-
immunosuppression. None of these patients received nal nucleated cells present and 203 mL (5 mL/kg) of
methotrexate, and none developed hemolysis. donor plasma. CFU-C recovery after plasma removal
averaged 95.2%.
TACROLIMUS (FK506) The degree of volume reduction that would prevent
hemolysis by a given antibody is difficult to deter-
Greeno and coworkers20 described a patient who mine, but some published information is pertinent to
developed no signs of hemolysis after a minor ABO- this point. Inwood and Zuliani,55 in reviewing a case
incompatible BMT after receiving methotrexate, anti- of hemolysis resulting from transfusion of a unit of
thymocyte globulin, and prednisone for GVHD group O packed RBCs to a group A person, deter-
prophylaxis. When her leukemia relapsed, however, mined that the group O unit contained anti-A1 activity
Immune Hemolysis Associated with Transplantation 469

to a titer of 8192. They estimated that the titer of the of the donor’s type. Because significant hemolysis
donor anti-A in the patient’s circulation after transfu- caused by antibodies in the donor marrow is rare and
sion was 164. This resulted in intravascular hemolysis can be prevented more easily by plasma reduction of
and a drop in hemoglobin of 2.4 g/dL. the donor marrow product when high-titer antibodies
Studies in bone marrow transplant recipients have are detected in the donor, it does not seem appropri-
indicated that transfusion of ABO-incompatible red ate to subject patients routinely to such an extensive
cells to patients with isohemagglutinin titers of 16 or procedure as exchange transfusion to avoid hemolysis
lower produced no evident hemolysis.37 Thus, in by passive transfer of antibody.
calculating the amount of plasma reduction that might We have performed pretransplant RBC exchange
be appropriate for a given product, if one reduces the transfusions for a group of patients receiving an
volume of plasma to be transfused so that the post- ABO-minor mismatched transplants from unrelated
transfusion isohemagglutinin titer will be lower than 16, donors and who received cyclosporine (but not
it appears likely that serious hemolysis will not occur. methotrexate) in the post-transplant period for
GVHD prophylaxis.12 This procedure was performed
after observing massive hemolysis in three consecu-
Plasma Reduction of Minor tive such patients. The amount of post-transplant
ABO-Incompatible Platelet Products hemolysis among subsequent patients was reduced
to Prevent Hemolytic Transfusion but not eliminated.
Reactions Sniecinski and O’Donnell59 have recommended pro-
phylactic RBC exchange of the transplant recipient for
One may reduce the volume of pooled platelet concen- all patients receiving a minor ABO-incomopatible stem
trates or plateletpheresis products to minimize risks cell product and post-transplant immunosuppression
that are related to the volume of plasma transfused.56,57 with cyclosporine without concurrent methotrexate.
Volume reduction has generally been recommended They further recommend that exchanges should be
for prevention of volume overload in infants, for carried out until 70%–80% of the patent’s RBC mass is
patients in danger of overload from intravenous fluids, replaced with group O RBCs. In contrast, Bolan and
patients with acute respiratory distress syndrome, and coworkers18 have concluded that a policy to perform
patients with congestive heart failure.57 Volume reduc- RBC exchanges prophylactically for all subjects at risk
tion might also be appropriate as a means of diminish- seems unwarranted. They determined that after a 10-
ing the amount of antibody that would be transfused. unit RBC exchange for a patient who weighed 90 kg,
Measuring the isohemagglutinin titers in all minor more than 40% of the recipient’s original RBC mass per-
ABO-incompatible platelet products is impractical sisted in circulation after the exchange.
and seems unnecessary, however, as episodes of Worel and colleagues39 also recommend prophy-
severe hemolysis due to platelet transfusions are rare. lactic RBC exchange, especially for patients who
Thus, the most practical policy is to try to avoid minor receive a PBSC graft in combination with a GVHD
ABO-incompatible platelet transfusions. If only minor prophylaxis regimen without methotrexate.
incompatible products are available, one must be Because pretransplant isohemagglutinin titers do not
cognizant of the possible consequences of passively appear to predict the incidence or severity of hemolysis
transfused antibody, which can effect compatibility
after minor ABO-mismatched transplants,59 selection of
testing and occasionally cause significant hemolysis
patients to undergo pretransplant exchange trans-
(see page 494).
fusion on the basis of these titers would not seem
An alternative approach for preventing hemolysis indicated.
is to wash platelets, which removes a large percentage
of the plasma. Pineda and associates,58 however,
reported that platelet recovery was decreased signi-
ficantly in patients receiving washed platelets. The Monitoring the Recipient
mean platelet recovery was 73% for unwashed for Development of Passenger
platelets, 23% for saline-washed platelets, and 52% for Lymphocyte Syndrome
ACD-saline–washed platelets. Platelet survival was Hemolysis resulting from passenger lymphocyte syn-
not significantly different among patients receiving drome usually has its onset between days 3 and
differently processed platelets, which indicates that a 15 after transplantation; therefore, one should rou-
fraction of the platelets remains undamaged and can tinely monitor all patients receiving a minor ABO-
survive normally. mismatched hematopoietic stem cell transplant for the
presence of hemolysis and for donor-derived iso-
Pretransplant RBC Exchange hemagglutinins during this period. A note in the
Transfusion medical record can serve to alert the personnel caring
for the patient regarding the appropriate procedures
One may avoid hemolysis caused by passive transfer and their rationales (Table 12-4). Usually, the syn-
of antibody in bone marrow products by performing drome is readily detectable by the presence of signs of
pretransplant RBC exchange transfusions using RBCs hemolysis, a positive DAT, and the relevant antibody
470 Immune Hemolytic Anemias

did not experience hemolysis. The researchers


TABLE 12-4. TRANSFUSION MEDICINE SERVICE concluded that serological findings—including the
RECOMMENDATIONS REGARDING ABO MINOR intensity of the DAT and the donor isohemagglutinin
MISMATCHED TRANSPLANTS titer—were not useful for predicting the occurrence of
severe hemolysis.
This patient has received an ABO minor mismatched transplant
(patient type , donor type and is therefore at
risk of developing delayed hemolysis from ABO blood group anti-
body transiently produced by donor “passenger lymphocytes” in the Management of Patients
transplanted organ, marrow, or peripheral stem cell product. with the Passenger Lymphocyte
“Passenger lymphocyte” hemolysis rarely appears before day 3 after Syndrome
transplantation, usually begins around day 7 to 10, and almost
always begins before day 15 if it is going to occur. It can cause a In most instances, hemolysis caused by the passen-
decrease in hemoglobin and hematocrit that can be misconstrued as ger lymphocyte syndrome can be managed by trans-
bleeding. Hemolysis is usually limited in intensity and duration but
can be quite serious in some patients.
fusion of compatible RBCs, the empirical use of
corticosteroids, avoidance of ABO-incompatible
We will follow the patient from day 3 to day 15 after transplantation
for the development of hemolysis, which can impact transfusion need plasma products, and maintenance of adequate renal
and blood selection. During this time, we recommend the following: perfusion. Such supportive care is generally ade-
Order daily hemoglobin/hematocrit, LDH, and total/indirect quate; hemolysis tends to be self-limiting because
bilirubin. newly formed red cells produced by the donor
Beginning on day 3, after transplantation, order a direct antiglo- marrow generally are not affected by the donor-
bulin test (DAT) three times per week and a reticulocyte count derived antibody.
two times per week (solid organ recipients) until day 15.
If there are any questions, please contact the Transfusion In those cases in which massive hemolysis occurs, it
Medicine Service. is reasonable to consider exchange transfusion, replac-
ing the patient’s antigen-positive erythrocytes with
group O RBCs. This therapeutic maneuver has been
used and perhaps is an effective means of preventing
(usually anti-A or anti-B) in the patient’s serum and in the renal failure that has resulted from hemolysis in
a RBC eluate. some patients.12 At least equally logical is plasma
We originally had expected that the most sensitive exchange transfusion, which should decrease the con-
test indicating the possible presence of hemolysis centration of the causative antibody. Hemolysis severe
would be the DAT and that serum antibodies would enough to require exchange transfusion is quite
always be readily detectable at the onset of hemoly- unusual, however.
sis. Experience, however, has indicted that in a
minority of cases, overt signs of hemolysis in the pas- Selection of Blood Products
senger lymphocyte syndrome might precede the
ability to detect the relevant antibodies on the In minor ABO-incompatible hematopoietic stem cell
patient’s RBCs or in the serum.16,20,23 Thus, in the transplants, it is generally most practical to transfuse
proper setting, the diagnosis of passenger lympho- group O red cells. This may be begun at the beginning
cyte syndrome must be accepted provisionally even of the preparative regimen. Although packed RBCs
without confirmatory serologic findings for 24–48 contain isoagglutinins reactive with the patient’s RBC,
hours. Accordingly, we pay particular attention to hemolysis caused by transfusion of plasma in packed
signs of hemolysis. The onset of the syndrome is RBCs is rare (see the foregoing discussion), so the use
often heralded by an abrupt drop in the patient’s of washed RBCs is generally unnecessary. (In those
hemoglobin and hematocrit associated with a dis- unusual cases in which the donor’s blood type is
tinct increase in serum bilirubin, especially of the group A or B and the recipient’s is group AB, the
indirect reacting type. Signs of intravascular hemol- donor’s type red cells may be used instead of group
ysis (hemoglobinemia and hemoglobinuria) also can O.) (Table 12-5)60
be present. Platelets and other plasma-containing products of
Bolen and associates18 found that a positive DAT recipient type are generally used to avoid the transfu-
was found a few days before or at the time of hemo- sion of isohemagglutinins that are reactive against red
lysis in their three patients who developed passenger cells of the patient, even though passive transfusion of
lymphocyte syndrome. On the other hand, a positive isohemagglutinins in plasma products is only infre-
DAT, including strong complement (C3d) coating of quently an important contributing factor. When the
RBC, also was detected in some patients who did not patient has converted to donor group RBC, ABO-
develop hemolysis. Also, serum isoagglutinins and matched platelets would be preferable, as ABO-
RBC eluates with specificity for the relevant ABO matched platelets are generally recommended for
antigen occasionally were detected in patients who patients receiving platelet transfusions.61
Immune Hemolysis Associated with Transplantation 471

TABLE 12-5. ABO-INCOMPATIBLE BONE MARROW TRANSPLANTATION (FROM


PREPARATIVE REGIMEN UNTIL ENGRAFTMENT)

Recipient Donor RBCs Platelets: First Choice Platelets: Second Choice* FFP*

Major Incompatible
O A O A AB, B, O A, AB
O B O B AB, A, O B, AB
A AB A AB A, B, O AB
B AB B AB B, A, O AB
O AB O AB A, B, O AB
Minor Incompatible
A O O A AB, B, O A, AB
B O O B AB, A, O B, AB
AB O O AB A, B, O AB
AB A A AB A, B, O AB
AB B B AB B, A, O AB
Major and Minor Incompatible
A B O AB A, B, O AB
B A O AB B, A, O AB

Note: Engraftment occurs when forward and reverse types are of donor and the DAT is negative.
* Avoid high titer ABO antibodies
From Friedberg RC: Transfusion therapy in the patient undergoing hematopoietic stem cell transplantation. Hematol Oncol Clin
North Am 1994;8:1105–1116.

PERSISTENCE OF BLOOD GROUP RBC Typing Irregularities after ABO


GLYCOSYLTRANSFERASE ACTIVITIES Minor Mismatched Hematopoietic Cell
AFTER HEMATOPOIETIC STEM CELL Transplants
TRANSPLANTATION Maeda and associates64 studied patients up to 72 weeks
after ABO-incompatible marrow transplants. They rec-
The carbohydrate structures of glycoproteins that ognized that isohemagglutinins that one would expect
confer blood group ABO specificities are synthesized to be produced by the donor marrow after BMT often
by glycosyltransferases, which are determined by were not detectable in the patient’s serum but, in some
genes located in different chromosomal loci. The patients, were found only on their RBCs. For example,
blood group A- or B-active structures are determined when marrow from a group A donor was used for
by the incorporation of N-acetyl-D-galactosamine or transplantation of a group B patient, one would expect
D-galactose onto a common acceptor carbohydrate that, after engraftment and replacement of the patient’s
chain (H substance) by the action of α-3-N-acetyl-D- group B RBCs with those of the group A marrow donor,
galactosaminyl transferases (A-glycosyltransferase) or one would find anti-B in the patient’s serum. Although
α-3-D-galactosyltransferases (B-glycosyltransferase). the expected isohemagglutinin was not found in the
Studies of plasma glycosyltransferase activities after serum, the DAT was positive, and anti-B could be
bone marrow transplantation have yielded fascinating eluted from the patient’s group A RBCs. Also, weakly
results. Matsue and coworkers62 reported that in eight positive reactions with anti-B were obtained when
patients transplanted with an ABO-incompatible testing the patient’s RBCs long after engraftment.
marrow, the ABO RBC type completely changed from The authors considered a number of possible expla-
the recipient type to the donor type but that preexistent nations for these findings, including the presence of
plasma glycosyltranserase activities of the recipient chronic mixed chimerism. They were unable to demon-
type persisted in seven of the eight patients. Weak strate mixed hematopoietic chimerism in the blood and
transferase activities of the donor type were observed bone marrow of their patients, however. They sug-
in all of the patients after marrow grafting. An example gested that the most likely explanation was that anti-B
of one such patient is illustrated in Figure 12-6. was produced by the donor marrow and reacted with
Yoshida and colleagues63 reported similar findings B antigen adsorbed from the patient’s plasma onto
in four patients. After BMT, the recipients’ A and B RBCs produced by the transplanted marrow cells.
enzyme activities changed slightly, and only weak Although routine ABO blood grouping after ABO-
reactivity of donor type was detected. Nevertheless, incompatible marrow transplantation generally indi-
the blood groups of the patients converted to those of cates conversion to donor type, we have observed
their bone marrow donors in each case (Table 12-6). weakly positive agglutination reactions a year or more
472 Immune Hemolytic Anemias

A RBCs FIGURE 12-6. Blood type and plasma N-


100 512 acetylgalactosaminyltransferase (A-enzyme) activity
B transferase
256 and galactosyltranferase (B-enzyme) activity before

Agglutination titer to anti-A or anti-B


and after bone marrow transplantation. The
80 128 patient, originally blood group B, received bone
marrow from a donor with blood group A. The
Group A or B RBCs (%)

64 percentage of RBCs was determined using a


60 32 microscope by counting agglutinated and
nonagglutinated cells, respectively. Transferase
16 activity was assayed after transforming group O
RBCs to A or B RBCs by incubation in the
40 8 presence of the patient’s plasma and UDP-N-
B RBCs A transferase
4 acetylgalatosamine (for A-tranferase) of UDO-
galactose (for B-transferase). The assay was
20 2 semiquantitated by titration using anti-A or anti-B
agglutinin with serial dilutions. (From Matsue K,
1
Yasue S, Matsuda T, et al: Plasma
0 0 glycosyltransferase activity after ABO-incompatible
bone marrow transplantation and development of
0 20 40 60 80 100 120 140 an inhibitor for glycosyltransferase activity. Exp
Days after BMT Hematol 1989;17:827–831.)

after marrow transplantation using anti-A,B sera when To determine whether mixed chimerism or adsorp-
testing RBCs from patients who had been group A tion of plasma antigen is responsible for these re-
before transplantation with a group O marrow (Branch actions, Arndt and coworkers67 performed flow
DR and Petz LD, unpublished observations).5 cytometry on RBCs of people with mixed cell popu-
These findings are consistent with reports of Renton lations, including patients who had been group A
and Hancock,65 who observed that when group O and who were transplanted with hematopoietic stem
RBCs are transfused to group A or B recipients, they cells from group O donors. They compared these
can acquire small amounts of A or B substance. The results with flow cytometry patterns obtained when
uptake of antigen is best demonstrated by using testing mixtures of group O and A RBCs that had a
certain group O sera and to a lesser extent with group small percentage of group A cells. Their results were
A and B sera. It can be demonstrated as early as four consistent with adsorption of group A and B sub-
days after transfusion and reaches a maximum after stance (rather than a mixture of group A and O
about 2 weeks (Table 12-7). Also, Crookston and RBCs) as the cause of the weakly positive agglutina-
Tilley66 have reported the adsorption of A antigen by tion reactions found in such post-transplant patients.
group O RBCs in group A1 marrow transplant recipi- These findings are discussed further with reference
ents who were transplanted with marrow from group to possible mechanisms of bystander immune
O donors. hemolysis (see Chapter 9).

TABLE 12-6. BLOOD GROUP AND PLASMA A- AND B-GLYCOSYLTRANSFERASE


ACTIVITIES BEFORE AND AFTER BONE MARROW TRANSPLANTATION

Patient Blood
Group Patient’s Plasma Enzyme Activity*
Patient Donor’s
Desig- Before After Blood Before BMT After BMT
nation BMT BMT Group A-Enzyme B-Enzyme A-Enzyme B-Enzyme

UPN 19 A1 O O 120 0 55–77 0


UPN 29 O A1 A1 0 0 0–27 0
UPN 35 O A1 A1 0 0 0–10 0
UPN 38 A1 B B 90–120 0 48–90 0–5

* Enzyme activity of the patient’s plasma was expressed as percentage of mean value of control plasma. Blood group
A- and B-enzyme activities are known to differ widely among individuals with phenotype A1 and B. A-enzyme activity in
nine plasma samples with phenotype A1 ranged from 2.8 to 16.8 (mean value 6.44), and B-enzyme activity in seven
plasma samples with phenotype B ranged from 5.0 to 17.5 (mean 10.6), when the activity was expressed as percent
of sugar transferred into fucosyllactose under the assay condition.
From Yoshida A, Schmidt GM, Blume KC, Beutler E: Plasma blood group glycosyltransferase activities after bone marrow
transplantation. Blood 1980;55:699–701.
Immune Hemolysis Associated with Transplantation 473

reported that ABO antibodies were not detected until


TABLE 12-7. AGGLUTINATION OF GROUP O RBCs more than 74 days after transplant in two patients
BY ANTI-A, ANTI-B AND ANTI-A,B AFTER transplanted with marrow from minor ABO-incom-
TRANSFUSION TO GROUP A OR B PATIENTS* patible donors. Gale and associates34 reported a
group O patient who was transplanted with marrow
Days after Average Score
from a group A donor who had IgM anti-B 2 years
Patient Group Transfusion O Sera A or B Sera
after BMT.
Mrs. H.D. A 2 0 0 Lee and coworkers69 reported that donor-derived
Mrs. A.S. B 4 0.2 0 ABO antibodies against recipient RBCs were detectable
Mrs. D.S. A 6 0.5 0 in five out of 36 patients who received minor ABO-
Mrs. D.S. A 13 0.9 0
Mrs. C.O. A 16 2.1 0.3
incompatible stem cell grafts. The probability of the
Mr. W.C. B 25 2.1 1.4 appearance of ABO antibodies by 1 year after trans-
Mrs. D.S. A 29 1.9 1.0 plant was 17.5%. The ABO antibodies appeared after
Mrs. D.S. A 48 1.4 0.3 the disappearance of recipient RBCs except in one
Mrs. H.D. A 49 1.5 0 patient, in whom there was no clinically detectable
Mr. W.C. B 76 2.4 0.9
hemolysis.
* Following transfusion of group A or B patients with group O RBCs, the
patient’s RBCs were treated with an excess of high titer anti-A or anti-B, as
appropriate, and the unagglutinated RBCs were separated from the
agglutinates by a centrifugal method. The free cells were washed and tested
MAJOR ABO BLOOD GROUP
with 20 to 30 fresh random group O sera and high-titer group A or B sera. INCOMPATIBLE HEMATOPOIETIC
Specificity of the agglutination observed was confirmed by the fact that
agglutination was inhibited by A or B secretor saliva but not with nonsecretor STEM CELL TRANSPLANTS
saliva or group O secretor saliva. Agglutination was not due to residual A or B
cells. The average score was calculated by adding together the scores for all
the sera and dividing by the number of sera tested. The average score gives Immune Hemolysis of Red Cells in the
an indication of the amount of antigen that the cells have taken up.
From Renton PH, Hancock JA: Uptake of A and B antigens by transfused group Stem Cell Product
O erythrocytes. Vox Sang 1962;7:33–38.
Because the volume of RBCs in a bone marrow
product could be equal to or greater than that in a unit
of RBC, the potential exists for an immediate
hemolytic transfusion reaction due to ABO incompat-
THE PRESENCE OF ibility. This may be prevented by removal of the RBCs
ABO ANTIBODIES LONG from the donor marrow or by removal of ABO anti-
AFTER MINOR ABO-INCOMPATIBLE bodies from the recipient. The latter procedure has
HEMATOPOIETIC STEM CELL sometimes been followed by pretransplant transfu-
TRANSPLANTATION sion of incompatible RBCs to adsorb the recipients’
hemagglutinins completely.
Antibody production by the donor marrow can be
present long after hematopoietic stem cell transplan- Delayed Hemolytic Reactions after
tation. For example, Lasky and colleagues54 reported Transfusion of Incompatible RBCs before
the presence of ABO antibodies more than 1 year Hematopoietic Cell Transplantation
after BMT in three of five recipients of minor ABO-
incompatible transplants who survived longer than The rebound of ABO antibodies after a transplant pro-
5 months after the transplant. The ABO antibodies cedure is of particular concern for patients who have
were first detected at 10 months (anti-A in AB recipi- been transfused with incompatible RBCs in an effort
ent, B donor) and at 21 and 27 months after BMT to further reduce the antibody titers by adsorption
(anti-A in A recipients, group O donors). Buckner and after plasma exchange. Such transfusions result in the
associates68 reported that six group A recipients of presence of a large volume of incompatible RBCs that
group O marrow transplants had anti-B (IgG titers of are susceptible to hemolysis when the antibody
16–128) more than 1 year after BMT but that none had rebound occurs. Indeed, Lasky and colleagues54 have
anti-A. Two other recipients of group O marrow had reported that delayed hemolysis occurred on days 6
blood type of group AB, and neither of these had to 10 after transplantation in seven of nine patients
detectable anti-B, but both had weak anti-A. who received incompatible RBC transfusions before
Although A antigen is known to be distributed the procedure. In two of these patients, the hemoglo-
widely in the tissues, no symptomatology could be bin dropped by 7.9 g/dL and 4.5 g/dL, respectively,
ascribed to the presence of the anti-A. Needs and over the course of 2 to 3 days. In contrast, none of five
coworkers33 reported on two group A2 patients who patients who had not received donor-type RBCs
were engrafted with group O marrow and produced developed delayed hemolysis. One of the transfused
anti-A1 after 24 months. Robertson and colleagues13 patients, who was group O, had pretransplant anti-A
474 Immune Hemolytic Anemias

titers of 32/128 (IgM/IgG) and anti-B titers of 64/16. incompatible BMT, this is not always the case (Fig.
These were reduced to undetectable levels by two- 12-7).71 Bensinger and colleagues37 pointed out that
plasma volume plasma exchanges followed by 4 units IgG antibody was detected for as long as 100 days
of incompatible, donor-type RBCs. On day 6 after the after a transplant procedure. This suggests the con-
transplant procedure, the ABO antibodies reappeared tinued production of IgG antibody by plasma cells
in the patient’s serum, signs of acute hemolysis that survived the pre-BMT cytoreductive therapy,
became evident, the DAT was positive for IgG and considering that the normal half-life of IgG is 21 days.
complement, anti-A and anti-B were eluted from the Other investigators have reported that isohemagglu-
patient’s RBCs, and the hematocrit dropped from 37% tinins were no longer detectable after 14 days to
to 18%.70 4 months after the transplant procedure.37,54,68,71,72
Buckner and associates68 reported on a patient who Prolonged persistence of ABO antibodies can
had been treated with transfusion of incompatible result in hemolysis of RBCs produced by the newly
blood before transplantation and in whom a rising anti- engrafted marrow. Sniecinski and associates 4
A titer after transplantation resulted in acute massive reported that anti-A or anti-B persisted for longer
intravascular hemolysis and renal failure. Although than 120 days after BMT in 9 of 58 evaluable patients
this was their only patient in whom hemolysis was a receiving a major ABO-incompatible marrow trans-
major factor in patient management, unwanted anti- plant. Five patients developed overt hemolysis at a
body reappeared in the serum of 12 of 14 evaluable time when antibodies were still detectable but had
patients. Bensinger and coworkers37 reported on a decreased to a low titer (≤4). Hemolysis started on
similar patient who received incompatible RBCs before days 37 to 105 after BMT (median = +65), persisted
the transplant procedure and who developed acute for 10–94 days (median = 36 days), and was mani-
hemolysis starting on the third day after transplanta- fested by a drop in hemoglobin of 1.5–4 g/dL
tion, leading to renal failure that required hemodialy- (median = 2.5 g/dL), increases in bilirubin and
sis. These investigators reported that they abandoned LDH, and decreases in serum haptoglobin. The five
as unnecessary the transfusion of incompatible blood cases of hemolysis occurred among 30 patients
for antibody absorption before transplantation. who received cyclosporine-prednisone for post-
transplant GVHD prophylaxis, and no episodes of
Hemolysis of RBCs Produced by Newly hemolysis occurred among 28 patients receiving
Engrafted Marrow Caused by both methotrexate and prednisone. Before the
decrease in the ABO antibody titers to low levels,
Persistence ABO Antibodies erythropoiesis was suppressed; after the antibodies
Although ABO antibodies usually become undetect- decreased to undetectable levels, hemolysis ceased
able during the second month after major ABO- and adequate erythropoiesis developed (Fig. 12-8).

1.0

0.8
Probability of detecting ABO antibodies

FIGURE 12-7. Probability of detecting host ABO


0.6 antibodies in 36 major ABO-incompatible marrow
receipients at varying times after transplantation.
(From Witherspoon RP, Storb R, Ochs HD, et al:
Recovery of antibody production in human
allogeneic marrow graft recipients: Influence of
0.4 time posttransplantation, the presence or
absence of chronic graft-versus-host disease,
and antithymocyte globulin treatment. Blood
1981;58:360–368.)

0.2

0.0
10 20 30 40 50 60 70 80 90 100 110 120
Days post-transplant
Immune Hemolysis Associated with Transplantation 475

UPN 355

Red cell transfusions

Hemolysis
256
Erythroid engraftment
128
FIGURE 12-8. Post-transplant course in a patient with no RBC
Platelet engraftment
production and high ABO antibody titers. Hemolysis occurred when 64
titers were low. Resolution coincided with undetectable antibodies.

Anti-A titers
Findings of hemolysis were present even though donor
32
marrow–derived RBCs were undetectable, suggesting
intramedullary hemolysis. Upper curve represents immunoglobin G
(IgG) ABO antibodies, and lower curve represents IgM ABO 16
antibodies. (From Sniecinski IJ, Oien L, Petz LD, et al:
Immunohematologic consequences of major ABO-mismatched bone 8 IgG
marrow transplantation. Transplantation 1988;45:530–534.)
4

2 IgM

20 40 60 80 100 120 140 160 180 200 220


Post-BMT days

Lopez and coworkers73 reported on a major ABO- and 30 evaluable patients, respectively, who were
incompatible BMT patient who developed hemolysis transplanted with major ABO-incompatible marrows.
between days 36 and 50 after transplant due to the
persistence of recipient isohemagglutinins.
Biggs and colleagues74 reported hemolysis of newly Suppression of Hematopoiesis by
formed group B RBCs occurring before the 33rd day Persistent ABO Antibodies after
after transplant in a group O patient transplanted Major ABO-Incompatible Hematopoietic
with a group B marrow. Cell Transplants
Transient hemolysis caused by immune hemolysis
by ABO antibodies of RBCs produced by the newly A number of investigators have reported impaired or
engrafted marrow seems to be observed infrequently. failed hematopoiesis in some patients with persistent
Neither Gmur and associates75 nor Bar and coworkers76 isohemagglutinins after BMT. Figure 12-9 illustrates
identified episodes of hemolysis in their reports of 15 the findings in a patient with a high titer anti-A at the

BMT

CMV
4096
2048 IgG
IgM
1024 Neut × 109/ l
FIGURE 12-9. Serial anti-A titers (reciprocal of
512 Retics × 1011/ l
dilution) and reticulocyte and neutrophil counts
during the weeks after BMT in a group O patient 256
transplanted with marrow from a group A donor.
128
Anti A–titers

(From Blacklock HA, Gilmore MJ, Prentice HG,


et al: ABO-incompatible bone-marrow 64 5
transplantation: Removal of red blood cells from
32 4
donor marrow avoiding recipient antibody
depletion. Lancet 1982;2:1061–1064.) 16 3
8 2
4 1
0 0
0 2 6 8 10 12 13 14 16 18
Weeks post-BMT
476 Immune Hemolytic Anemias

13 Plasma exchanges
UPN 279
2048
1024
lgG ABO antibody titers

512
256
128 UPN 334 FIGURE 12-10. IgG-ABO antibody titers in five
patients whose antibodies persisted for >255
64 days. (From Sniecinski IJ, Oien L, Petz LD, et al:
UPN 344
32 Immunohematologic consequences of major
ABO-mismatched bone marrow transplantation.
16 UPN 320 Transplantation 1988;45:530–534.)
8
4 UPN 108
2

20 60 100 140 180 220 260 300 340 380


Days post BMT

time of transplant, who had detectable antibody until Sniecinski and colleagues4 reported that, in 9 of
15 weeks after BMT. She had delayed engraftment of 58 evaluable patients who received a major ABO-
red cells, neutrophils, and platelets (the latter is not incompatible BMT from an HLA-matched sibling
illustrated).77 donor, there was persistence of isohemagglutinins for
more than 120 days after BMT. Red cell production
was delayed to 40 days or longer in the nine patients
Pure Red Cell Aplasia and, in five of these, erythropoiesis was markedly
Much more common than suppression of hemato- delayed to 170 days or longer (Fig. 12-10).
poiesis is a delay in the production of donor-type Gmur and associates75 reported on three patients
RBCs—a temporary development of pure RBC who developed PRCA lasting from 5 to 8 months.
aplasia (PRCA) until the titers of isohemagglutinins One patient’s course is illustrated in Figure 12-11. Bar
reach low levels.1,4,37,54,59,69,75,76,78-84 This can be and coworkers76 reported that 8 of 30 patients (27%)
explained by the interaction of anti-A or anti-B with with major ABO incompatibility had lymphocyte-
donor erythroid precursors expressing the A and/or depleted bone marrow with no detectable donor
B antigens.85-87 RBCs for at least 2 months after BMT. In these

250

1024 200
Reticulocytes/mm ( )

512
FIGURE 12-11. Course of anti-A agglutinin
256
150 titer (squares) and absolute reticulocytes/
Titer ( )

128 mm (open circles) after ABO-incompatible


BMT in UPN 39. Day 0 is the day of BMT.
64
Arrows indicate plasma exchange. (From
32 100 Gmur J, Burger J, Schaffner A, et al: Pure
red cell aplasia of long duration
16
complicating major ABO-incompatible bone
8 marrow transplantation. Blood
50 1990;75:290–295.)
4
2
0 0

0 60 120 180 240 300 360


Day
Immune Hemolysis Associated with Transplantation 477

patients, there was an early rise in or a persistence of A 100


high isohemagglutinin titers (IgG more than IgM),
whereas the titers decreased immediately after BMT NST

% Without donor RBC


80
in patients without delayed erythropoiesis. The
median pretransplant IgG isohemagglutinin titer

chimerism
60
(anti-A or anti-B) was 16 (range, 1–512) in patients
without delayed erythropoiesis, in contrast to a
40
median titer of 512 (range, 64–16,000) in patients with
a delayed onset of erythropoiesis. The IgM iso-
hemagglutinin titers were the same in both groups. 20
Myeloablative
Various reports in the literature do not agree on a SCT
correlation between pretransplant ABO titers and the 0
number of days after transplantation before the onset 0 25 50 75 100 125 150 175 200 225
of erythropoiesis.76
Lee and colleagues69 analyzed data for 40 patients
B 100
with major (and/or minor) ABO incompatibility and
reported that recipient-derived ABO antibodies

isohemagglutinin > 1+
against donor-type RBCs had disappeared by 80

% with anti-donor
(median) day 89 after the transplant procedure NST
(range, 25–449 days); the probability of ABO anti- 60
body disappearance by 1 year after BMT was 97.3%.
ABO antibodies against donor-derived RBCs dis- 40
appeared more rapidly after unrelated donor trans-
plantation (P = 0.006) and in patients with acute 20
GVHD (P = 0.025). Out of 35 patients, 10 (28.6%) evi- Myeloablative
denced persistent reticulocytopenia for more than 60 SCT
0
days after transplantation.
0 25 50 75 100 125 150 175 200 225
As with other immunohematologic complications
after BMT, the incidence of delayed hematopoiesis Time after transplantation (days)
appears more common among patients who were FIGURE 12-12. The onset of donor RBC chimerism and decline in
treated with cyclosporine for post-transplant GVHD antidonor isohemagglutinin levels after NST compared with
prophylaxis.4,59,75,76 myeloablative SCT. The onset of donor RBC chimerism and decline of
host antidonor isohemagglutinin to clinically insignificant levels were
delayed markedly following major ABO-incompatible NST compared
PRCA after Reduced-Intensity with myeloblative SCT. NST data are represented by solid squares;
myeloblative SCT data, by open circles. SCT, stem cell transplant; NST,
Nonmyeloablative Conditioning Regimens nonmyeloablative stem cell transplant. (A) Kaplan-Meier plot of the
percentage of transplantation. The time until detection of donor RBC
Bolan and associates79 determined PRCA to be present chimerism was significantly prolonged following NST vs. myeloblative
when bone marrow biopsy demonstrated adequate SCT; P < .0001. (B) Kaplan-Meier plot showing the percentage of
myeloid, lymphoid, and megakaryocyte populations in patients with persistent host antidonor isohemagglutinins greater that
the setting of absent or nearly absent erythroid precur- 1+ in strength. Isohemagglutinins decreased significantly faster after
myeloblative SCT than after NST; P = .012. (From Bolan CD, Leitman
sors and profound peripheral blood reticulocytopenia. SF, Griffith LM, Wesley RA, Procter JL, Stroncek DF, et al: Delayed
The found that PRCA occurred in four of 14 patients donor red cell chimerism and pure red cell aplasia following major
(29%) after hematopoietic stem cell transplants using ABO-incompatible nonmyeloablative hematopoietic stem cell
reduced-intensity nonmyeloablative regimens, but in transplantation. Blood 2001;98:1687–1694.)
none of 12 patients transplanted using myeloabative
conditioning during the same period of time. Donor
RBC chimerism (initial detection of donor RBCs in Peggs and coworkers88 reviewed their results with
peripheral blood) was delayed markedly after non- 15 major or bidirectional ABO-incompatible trans-
myeloablative regimens (median, 114 days vs. 40 days; plants done with nonmyeloalative conditioning regi-
P < 0.0001) and correlated strongly with decreasing mens. They concluded that the incidence of delayed
host anti-donor ABO antibody levels (Figs. 12-12 and donor red cell chimerism and PRCA likely differs not
12-13). They pointed out that strategies to further only between myeloablative and nonmyeloablative
reduce transplant-related toxicity, such as lower-inten- conditioning regimens but also according to the type
sity conditioning and more prolonged administration of nonmyeloablative (or reduced-intensity) condition-
of GVHD prophylaxis, could also be associated with ing regimen and perhaps also to the degree of B-cell
prolonged host ABO antibody production and could suppression achieved. PRCA after major ABO-incom-
result in marked delays in the onset of donor patible reduced-intensity transplantation has also
erythropoiesis. been reported by Veelken and colleagues.89
478 Immune Hemolytic Anemias

A 250 transplantation or cryopreservation to reduce the


amount of hemoglobin transfused with the product.59
200
Red Cell Removal from Donor Marrow
RBC chimerism (d)
Onset of donor

150
The main objective of removing RBCs from the donor
marrow is to remove the majority of RBCs while pre-
100 serving the hematopoietic progenitors to encourage
R 2 = 0.7519
engraftment. Several manual and automated tech-
50 niques have been developed to remove RBCs selec-
tively. Some techniques isolate the mononuclear
0 cell-rich component where the cells necessary for
0 50 100 150 200 250 engraftment are found, whereas others use the entire
Time to host antidonor ABO antibodies ≤ 1+ (d) buffy coat that contains both mononuclear and poly-
morphonuclear cells. These techniques have been
reviewed by Sniecinski and coworkers.59
B 250
The major risks of RBC depletion from marrow
include stem cell loss during processing and the
hazard of infusing small amounts of incompatible
isohemagglutinin ≤ 1+ (d)

200
Time to host anti-donor

RBCs. Dinsmore and associates90 reported that two


150 patients who received 68 mL and 45 mL of red cells,
respectively, developed transient hemoglobinuria
without renal impairment. Warkentin and cowork-
100
R 2 = 0.5638 ers91 reported that their marrow products contained
0.4–21.8 mL of red cells but that even the transfusion
50 of these small volumes of red cells resulted in mea-
surable evidence of hemolysis in some patients,
0 although reactions were not severe. Braine and col-
0 10 100 1000 10000 leagues,92 whose protocol left 8–38 mL of incompat-
Pretransplantation host antidonor ABO antibody titer ible erythrocytes in the infusate, reported somewhat
FIGURE 12-13. Correlation of the onset of donor RBC chimerism with a more severe reactions, including fever, hypertension,
decline in host antidonor ABO antibodies after major ABO-incompatible chills, hemoglobinuria, bradycardia, and confusion.
nonmyeloablative stem cell transplant (NST). The onset of donor RBC The total volume of RBCs in the marrow product
chimerism was strongly correlated with a decline in host antidonor ABO that is infused might determine the frequency of
antibodies to 1+ or lower, and the time until ABO antibodies decreased
to levels 1+ or lower was related to the pretransplantation ABO antibody adverse reactions; several authors who reported
titer. (A) The onset of donor RBC chimerism after NST strongly maximum volumes of residual RBCs of 10 mL or less
correlated with a decline in host isohemagglutinins to clinically stated that the infusions caused no clinical symptoms
insignificant levels (1+ or lower on reverse type; R2 = 0.72, P < 0.0005). or evidence of hemoysis.77,93-95 It is also likely that the
(B) The time until host antidonor ABO antibodies decreased to levels
1 + or lower correlated with the log value of the pretransplantation ABO
titer of the patient’s ABO antibodies is of significance,
antibody titer (R2 = 0.56; P = 0.02). (From Bolan CD, Leitman SF, Griffith although this factor has not been studied extensively
LM, et al: Delayed donor red cell chimerism and pure red cell aplasia in this setting.
following major ABO-incompatible nonmyeloablative hematopoietic stem
cell transplantation. Blood 2001;98:1687–1694.)
Removal of ABO Antibodies
from the Patient
MANAGEMENT OF PATIENTS The purpose of this approach is to decrease the titer
of the potentially offending antibody before marrow
RECEIVING A HEMATOPOIETIC STEM infusion. This may be achieved by the use of plasma
CELL TRANSPLANT FROM A MAJOR exchange or plasma immunoadsorption, usually
ABO-INCOMPATIBLE DONOR carried out daily over the course of 3 to 4 days before
marrow infusion. The goal is to reduce the antibody
Prevention of Acute Hemolysis titers to 16 or less, as transfusion of marrow products
to patients with such low titers have been reported
Two general principles are used to prevent acute to not result in acute hemolytic reactions. 15,37
hemolysis resulting from transfusion of incompatible Supplementation of this procedure by transfusion of
RBCs at the time of donor marrow infusion. One can incompatible RBCs is not recommended because of
either remove RBCs from donor marrow products or the risk of delayed hemolysis caused by antibody
remove isohemagglutinins from the patient. In an rebound in the post-transplant period.
analogous fashion, one should minimize the RBC Rowley15 states that up to 4 days of plasma
content of peripheral blood stem cell products before exchange might be required (1 day for patients with
Immune Hemolysis Associated with Transplantation 479

antibody titers of 32–128, two days for titers of donors are used, should undergo some form of anti-
256–512, three days for a titer of 1024, and four days for body depletion—perhaps before marrow infusion, but
a titer of 2048). Plasma exchange is unlikely to be ade- particularly after transplantation—if marrow devel-
quate for the management of patients with titers opment is not proceeding. They indicate, however,
greater than 2048; these patients should receive red that donor marrow red cell depletion alone is proba-
cell–depleted products. bly a sufficient measure for the majority of ABO-
Disadvantages of methods to remove antibody incompatible transplants, especially among recipients
from the patient are that the procedures are lengthy who have low anti-A or anti-B titers.
and need to be carried out during the period immedi- Patients who have high isohemagglutinin titers
ately before transplantation, when the patient is being before transplantation should be followed with
conditioned for transplantation with irradiation and weekly titers after transplantation. A rising titer
chemotherapy.1,96 Also, the patient is exposed to the usually preceeds the onset of delayed erythropoiesis,
risks of transfusion, including allergic reactions and hemolysis, or both.59 Plasma exchange or immuno-
the transmission of infectious diseases. Finally, in a adsorption should then be considered. Lee and associ-
considerable number of patients, the antibody titers ates69 also reported that 7 of 12 patients who showed
rise again after transplantation (Table 12-8).37,78,96-98 a post-transplant increase in titers of isoagglutinins
against donor-type RBCs developed delayed erythro-
poiesis, whereas only 3 of 23 patients who did not
Prevention of Delayed Erythropoiesis show a post-transplant increase in the isoagglutinin
Some investigators recommend that even if RBCs are titers developed red cell aplasia.
removed from the donor marrow product, removal of Reviron and coworkers99,100 suggested that it might
the patient’s isohemagglutinins by plasma exchange be appropriate to remove isohemagglutinins from the
should be carried out before transplantation. This rec- patient on day 0 if the titer of antibody is 512 or
ommendation is based on the expectation that reci- higher. They did not present data, however, to indi-
pients who have isohemagglutinins reactive against cate that outcomes are improved when this strategy is
donor blood group antigens—particularly if they are chosen compared with a policy of waiting to perform
in high titer—might experience a longer delay in the plasma exchange until after transplantation for
onset of erythropoiesis than those who have the iso- patients in whom the isohemagglutinin titers do not
hemagglutinins removed. Observations about this diminish spontaneously. They suggest that day 20
point have been in disagreement. Jin and associates95 after transplantation is an appropriate time to evalu-
found no difference in erythrocyte transfusion require- ate a patient to determine whether plasma exchange
ments between recipients of a red cell–depleted graft or immunoadsorption should be performed.
and patients who had plasma exchange or immuno-
adsorption, whereas Lasky and coworkers54 found Treatment of Delayed Erythropoiesis
that red cell transfusions were required in significantly
larger numbers and for a longer time among recipients Appropriate management of patients with a delayed
of red cell–depleted marrow than among patients pre- onset of erythropoiesis is not clear.15,79 Treatment suc-
pared by plasma exchange. cesses and failures in PRCA after hematopoietic stem
Blacklock and colleagues77,85 recommend that reci- cell transplantation have been described with plasma
pients with high-titer IgG anti-A, especially when A1 exchange4,101–103 and erythropoietin.73,81,104–106 A
patient described by Ohashi and colleagues104 did not
respond to erythropoietin alone, but erythropoiesis
began after the addition of methylprednisolone.
TABLE 12-8. SERIAL ANTI-A TITERS FOLLOWING This development could have been a coincidence,
PLASMA EXCHANGES AND INFUSION however, as the rise in reticulocytes in this patient was
OF A SUBSTANCE accompanied by a drop in anti-A titer to undetectable
levels. Intravenous immune globulin was not found to
Time IgM IgG be beneficial in two case reports; in two other cases,
antilymphocyte globulin did result in restoration of
Baseline 2048 512
Postplasma exchange-1 (10 L) 64 32
erythropoiesis.82,102
Post-A-substance 64 32 For most patients, erythroid engraftment occurs
Preplasma exchange-2 64 32 spontaneously within 6 months when isohemagglu-
Postplasma exchange-2 (18 L) 1 2 tinin titers decrease to low levels (generally ≤4).
Transplant 128 64 Indeed, Benjamin and associates83 indicated that
Day +5 2048 64
Day +12 2048 64 PRCA rarely requires intervention other than transfu-
Day +19 512 16 sion support. RBC production can be suppressed for
Day +35 0 0 more than a year, and in one patient, the onset of ery-
Day +48 0 0 thropoiesis was delayed until 5 years after transplan-
From Hershko C, Gale RP, Ho W, Fitchen J: ABH antigens and bone marrow tation,107 which was long after corticosteroids and
transplantation. Br J Haematol 1980;44:65–73. multiple plasma exchanges had failed to produce
480 Immune Hemolytic Anemias

benefit.4,107 Occasionally, isohemagglutinins might poietin (Fig. 12-14). A 25-year-old male with chronic
still be present at titers as high as 16 when the red cell phase chronic myeloid leukemia (CML) underwent a
aplasia resolves.4 BMT from his HLA-identical sister. The patient was
Other reports have documented resolution of PRCA group O, and the donor was group A. The patient
after donor lymphocyte infusion.108,109 Bolan and received erythropoietin on days 0 through 36 after
coworkers79 reported that erythropoietin was ineffec- transplantation because he was included in a prospec-
tive in all four of their patients, but that discontinua- tive randomized protocol to test the usefulness of ery-
tion of cyclosporine appeared to lead to resolution of thropoietin in BMT. Anti-A was detected persistently in
PRCA. Similarly, Yamaguchi and colleagues110 the patient’s serum throughout the post-transplant
described a patient who needed RBC transfusion course until day 175. Between days 36 and 50 after
every week from day 54 after hematopoietic cell transplantation, his hemoglobin dropped from 12.8
transplantation onward. He showed no evidence of g/dL to 9.7 g/dL. On day 50, his DAT was positive
GVHD, but with the tapering of cyclosporin after day with anti-C3, anti-A was eluted from his RBC, the LDH
123, he developed chronic GVHD around day 145. that had been 381 IU/L on day 36 was now 860 IU/L
The patient no longer needed transfusion from day (normal = <360 IU/L), and the serum haptoglobin
167, the reticulocyte count began to increase on day value was 0. Administration of erythropoietin was
179, and antidonor isohemagglutinin titers became resumed, and reticulocytes rose to as high as 90%. Signs
undetectable. They concluded that chronic GVHD of hemolysis persisted, and the hemoglobin remained
induced by tapering of cyclosporin appeared to be stable until after day 78. Thereafter, signs of hemolysis
related to improvement in the PRCA. A graft-vs.- gradually subsided, the hemoglobin improved, and
plasma cell effect is consistent with the data of Lee erythropoietin was tapered and then discontinued on
and associates,69 cited previously, and of Mielcarek day 150. The patient’s anti-A titers on day 141 were IgM
and coworkers.111 8 and IgG 16. In this patient, erythropoietin might have
caused release of RBCs by the newly engrafted marrow
earlier than would have otherwise occurred in the face
Treatment of Delayed Hemolysis of persistent anti-A, thus minimizing the period of red
Management of this rather uncommon complication cell aplasia but at the cost of hemolysis, which in this
of major ABO-incompatible transplants has required instance caused no adverse consequences.
only supportive management with transfusion of
group O RBCs. Signs of hemolysis of newly formed Selection of Blood Products
RBCs can persist for 10–94 days (median = 36 days).
Lopez and colleagues73 reported on a patient with From the onset of the preparative regimen, it is
delayed hemolysis whose treatment included erythro- advisable to use group O RBCs when transfusion is

EPO doses

16 Reticulocytes Hemoglobin
14
Hemoglobin (g/dL)

12
10
100
FIGURE 12-14. Top: Response of
Reticulocytes %

8
75 hemoglobin and reticulocytes to EPO
6 administration. Bottom: Serum EPO
50 levels and parameters of hemolysis
4
before and after EPO therapy. N.T., not
2 25 tested. (From Lopez J, Steegmann JL,
Perez G, et al: Erythopoietin in the
0
0 treatment of delayed immune hemolysis
36 50 64 78 102 116 130
of a major ABO-incompatible bone
Days post-BMT marrow transplant. Am J Hematol
Normal range 1994;45:237.)
LDH (U/L) 381 860 751 574 451 419 347 <360

Bilirubin (mg/dL) 1.2 N.T. 0.9 N.T. 0.7 0.7 0.6 0.3–1.3
Haptoglobin (U/L) N.T. 0 0 0 N.T. 106 145 50–150

Serum EPO 128 28 186 N.T. N.T. 156 310 4–28


levels (mU/mL)
Immune Hemolysis Associated with Transplantation 481

necessary, although the recipient’s RBC type can be 100 Control

Cumulative alloimmune platelet


used for group A or B patients in those unusual cases UVB-PC
in which the donor is group AB (see Table 12-5). 80 F-PC

refractoriness (%)
Ultimately, when the patient’s hemagglutinins have F-AP
become undetectable, red cells of the donor’s type 60
may be transfused.
Also, it is reasonable to minimize the administra- 40
tion of plasma that contains isohemagglutinins that
will react with RBCs of the donor’s type. This is best
20
accomplished by providing platelet products that are
of the donor’s type. If such platelets are not available,
volume reduction of the platelets may be performed, 0
although the amount of antibody in the platelet prod- 0 2 4 6 8
ucts is usually not a significant factor in causation of Weeks
hemolysis. The use of washed RBCs or washed FIGURE 12-15. Development of alloimmune platelet refractoriness in
platelets as a means of minimizing plasma adminis- patients who had refractoriness and also were antibody-positive. (From
tration is generally unnecessary. Leukocyte reduction and ultraviolet B irradiation of platelets to prevent
alloimmunization and refractoriness to platelet transfusions. The Trial
to Reduce Alloimmunization and Refractoriness to Platelets Study
Group. N Engl J Med 1997;337:1861–1869.)
Other Considerations in Selection
of Blood Products
Although the following consideration is not pertinent THE ROLE OF THE ABO BLOOD GROUP
to the topic of immune hemolysis, one must be aware SYSTEM IN GRAFT-VS.-HOST DISEASE,
that all cellular blood products should be γ-irradiated RELAPSE, AND MORTALITY AFTER
for all hematopoietic stem cell transplant patients ALLOGENEIC HEMATOPOIETIC CELL
beginning with the onset of the preparative regimen.
Friedberg112 also suggests that, for patients who are to TRANSPLANTATION
have a stem cell transplant, blood products intended
for transfusion that might still be circulating at the Graft-vs.-host disease (GVHD) is most commonly man-
time of stem cell collection should be γ-irradiated. ifested by a generalized maculopapular rash, hepatitis,
One unresolved question concerns the duration of diarrhea, and a delayed reconstitution of hematopoietic
time after transplantation during which irradiated and lymphoid function.114 GVHD is a pathologic
products should be administered. It seems logical to process initiated by engrafted immunocompetent
extend their use during the period of posttransplant donor T-lymphocytes responding to alloantigens
immune deficiency. This can persist for 1 or 2 years expressed on host cells, particularly cells derived from
and might be present even longer in patients who the lymphohematopoietic system. As the role of ABO
have GVHD. A common policy is to irradiate cellular incompatibility was being evaluated early in the
blood products indefinitely in patients who have history of BMT, there was concern that minor ABO
received a hematopoietic stem cell transplant.112 blood group incompatibility might be associated with a
Also, cytomegalovirus (CMV) transmission should higher incidence or greater severity of GVHD.
be minimized by the use of appropriate CMV-safe pro- In an early report, Storb and associates115 stated that
ducts. The indications for CMV-safe products and the their experience did not support the concept that ABO
choice of products are beyond the scope of this text. antigens played a role in causing GVHD, as only one
Leukocyte-reduced products are generally given to of ten patients who received a minor ABO mis-
minimize refractoriness to platelet transfusion. A multi- matched marrow developed GVHD.
institutional, randomized, blinded study of alloimmu- Buckner and coworkers68 analyzed the results of
nization to platelets determined that without the use of BMT with regard to the incidence and severity of
leukocyte-reduced blood products, 13% of previously GVHD among 39 evaluable patients with aplastic
untreated patients treated for acute myelogenous anemia or acute leukemia who had minor ABO mis-
leukemia became alloimmunized and refractory to matches with their donors. Only patients who were
platelet transfusions.113 With the use of leukocyte- genotypically HLA-matched at the A, B, and D loci
reduced products, 3–4% became alloimmunized and with a sibling donor were included in the analysis.
refractory (Fig. 12-15). The study also indicated that There were 229 evaluable patients with ABO-identical
patients who had detectable lymphocytotoxic antibod- donors available for comparison. Their results sug-
ies at entry into the study did not benefit from transfu- gested no effect of minor ABO incompatibility on the
sion of leukocyte-reduced platelets. Finally, platelets incidence or severity of GVHD.
obtained by apheresis from single random donors pro- Hershko and colleagues98 extended earlier observa-
vided no additional benefit compared with pooled tions34 and reported a comparison of the incidence of
platelet concentrates from random donors. GVHD among 60 patients with ABO-identical or major
482 Immune Hemolytic Anemias

mismatched grafts with the incidence of GVHD among Stussi and associates119 evaluated the effect of ABO
15 patients with minor ABO-mismatched marrow trans- incompatibility on GVHD in 173 consecutive patients
plants. There was no statistically significant difference receiving allogeneic bone marrow transplants.
between the two groups in the incidence of GVHD. Univariate analysis suggested a higher incidence of
Lasky and associates54 found no statistical differ- GVHD in minor ABO incompatibility than in ABO
ence in the incidence of GVHD when comparing 13 identity (14/30, 47% vs. 37/112, 33%; P = 0.02). Using
patients who received a minor ABO-mismatched BMT logistic regression adjusted for potential confounders,
with 95 patients who received ABO-matched marrow however, the risk of GVHD did not differ significantly
grafts. Other investigators have reached similar between the two groups. In a further report, Stussi and
conclusions.77,91,116 coworkers120 reported the results of a retrospective
On the other hand, Bacigalupo and coworkers117 two-center analysis of 562 consecutive patients receiv-
reported an analysis of 174 patients who received a ing allogeneic bone marrow or peripheral blood stem
BMT from an HLA-identical sibling for severe aplastic cell transplantation. The incidence of acute GVHD
anemia, acute lymphoblastic leukemia, or chronic gran- (grades I–IV) was higher for minor ABO-incompatible
ulocytic leukemia. Twenty-three transplants were ABO transplants compared with transplants involving ABO
major-mismatched, 27 were ABO minor-mismatched, identity (relative risk [RR], 2.8; 95% confidence interval
and 124 were ABO-matched. Double mismatched [CI], 1.3–5.9; P = 0.009). This difference was limited to
grafts (A to B and B to A) were excluded from their mild GVHD; in moderate-to-severe GVHD (grades
analysis. They concluded that ABO compatibility II–IV), no significant difference in incidence was found
clearly was associated with a different risk of grade among the groups. Other findings were that RBC
II–IV acute GVHD (Fig. 12-16). Recipients of ABO engraftment was delayed in major ABO-incompatible
minor-mismatched grafts experienced the highest inci- transplants and that the relapse rate was not influenced
dence of GVHD, recipients of ABO-identical grafts had by ABO incompatibility.
an intermediate risk, and patients given an ABO major-
mismatched BMT had the lowest incidence. To explain
the low incidence of GVHD in ABO major-mismatched
Transplant Outcome
BMT, the authors suggested that donor lymphocytes Benjamin and colleagues121 performed a retrospective
could absorb ABO antigens and then become the target analysis of a cohort of 292 allogeneic transplant
for immune destruction by anti-A/B antibodies, thus recipients and measured survival in a subgroup of
contributing to a low incidence of GVHD. ABO-incompatible bone marrow graft recipients.
Barone and colleagues118 retrospectively studied 65 They found that patients with acute myelogenous
bone marrow transplant patients who had ABO incom- leukemia or myelodysplastic syndrome receiving ABO-
patibility (major or minor) and compared the results incompatible non-T-cell–depleted marrow grafts had
with a control group whose donors were ABO- and Rh- an 85% greater risk of death within 100 days of trans-
compatible. They found GVHD in 26 patients (40%) plant (RR, 1.85; 95% CI, 1.33–2.58; P = 0.003) than com-
with major ABO incompatibility and in 15 patients parable patients receiving ABO-compatible grafts. Both
(23%) with minor incompatibility. They suggested that ABO major- and minor-mismatched graft recipients
GVHD frequency was increased in the group with ABO were at risk. The increased mortality rate was not due
blood system incompatibility compared with their to an increase in graft failure or acute GVHD; rather,
control group and the data in the literature. patients died of multiple-organ failure and sepsis, con-

100

90 ABO minor =/ 27 pts.


82%
80
% of pts. with acute CvIID

70
FIGURE 12-16. Occurence of acute GVHD in patients stratified
60 ABO = 124 pts. 54% according to donor-recipient ABO compatibility. Recipients of ABO
50 minor-mismatched grafts show a significantly (P = 0.01) higher
39% incidence of acute GVHD compared with recipients of ABO-
ABO major =/ 23 pts.
40 matched and ABO major-mismatched grafts. (From Benjamin RJ,
McGurk S, Ralston MS, Churchill WH, Antin JH: ABO incompatibility
30 as an adverse risk factor for survival after allogeneic bone marrow
transplantation. Transfusion 1999;39:179–187.)
20

10

0 18 36 54 72 90 108
Days post-BMT
Immune Hemolysis Associated with Transplantation 483

sistent with regimen-related toxicity. This effect was not by the donor’s immune system against antigens on red
seen in a larger group of 112 chronic myelogenous cells of donor origin, thereby qualifying these episodes
leukemia patients undergoing similar treatment. of hemolytic anemia as AIHA. The source of the
In the earlier study by Stussi and associates,119 the autoantibodies has not been documented definitively
authors found that survival was significantly depend- by immunoglobulin allotyping, however.
ent on ABO-compatibility (P = 0.004). In particular, Klumpp and associates128 reported on a patient who
patients with bidirectional ABO-incompatibility had an developed AIHA 19 months after BMT for chronic
excess mortality rate (RR, 7.6; 95% CI, 2.5–23.2; myelogenous leukemia using her HLA-identical,
P = 0.0004). They suggested that the role of ABO incom- ABO-identical brother as the donor. After a rather
patibility in allogeneic stem cell transplantation should uneventful clinical course for 18 months, the patient
be reevaluated in larger trials. In their subsequent experienced a drop in hematocrit from 30–35% to 21%
report, they performed a multivariate analysis adjusted associated with reticulocytosis (7.8%), mildly elevated
for potential confounders and found that survival was serum bilirubin and LDH, and decreased serum hapto-
significantly associated with ABO incompatibility globin. A DAT was positive, and a warm-reacting
(P = 0.006).120 Compared with ABO-identical trans- panspecific IgG and a cold-reacting IgM antibody were
plants, bidirectional ABO incompatibility increased the demonstrated. The patient also developed leukopenia
risk of mortality significantly (RR, 2.8; 95% CI 1.5–5.1; and thrombocytopenia, and an assay for neutrophil
P = 0.009), whereas survival rates for patients with antibody was positive, but platelet antibody could not
minor or major ABO-incompatible transplants were not be demonstrated. Numerous tests for chimerism using
significantly different from one another. chromosome analysis, Southern blot hybridization
Similar data were reported by Worel and col- using a bcr probe, and restriction fragment length
leagues121a who evaluated 40 patients transplanted polymorphism revealed no evidence of residual reci-
after nonmyeloablative conditioning. Eleven patients pient hematopoiesis or lymphopoiesis. Accordingly,
received a minor or bidirectional ABO-mismatched the antibodies were thought to arise as a result of a
graft, and eight received a major ABO-mismatched donor-antidonor reaction.
graft. Significantly more patients with ABO mis- Bashey and coworkers129 reported on a 17-year-old
matched grafts showed transplant-associated compli- male who was transplanted with marrow from his
cations and died as a result of transplant-related causes. HLA-identical brother; the patient and donor were
In contrast, a number of other studies did not find both group O with Rh phenotype CDce and probable
improved relapse-free survival associated with ABO- Rh genotype R1r. After day 170, he developed pancy-
compatible-vs.-incompatible transplants.37,54,111,122-125 topenia with hemoglobin at 6.6 g/dL, platelets at
In particular, Mielcarek and coworkers111,126 analyzed 55,000/μL, and WBC count at 1000/μL. Hemolysis
outcomes of 921 patients with hematologic malignan- was suggested by elevated reticulocyte count (11%),
cies transplanted between 1983 and 1998 from elevated total serum bilirubin (30 μmol/L), and ery-
HLA-identical–related donors after myeloablative throid hyperplasia of the marrow. The DAT was
conditioning. In this relatively homogeneous patient strongly reactive using anti-IgG and anti-C3d sera. The
population, presence of minor, major, or bidirectional patient’s serum contained antibody reacting weakly by
ABO incompatibilities did not affect outcome, and lysis and agglutination of papain-treated test cells, and
mismatching was not associated with decreased by the IAT using anti-IgG antiglobulin serum. The red
relapse rates or improved survival. cell eluate was strongly reactive in the IAT, and
Badros and colleagues127 performed a retrospective autoantibodies with various specificities within the Rh
analysis of the effects of ABO incompatibility among system were demonstrated in the serum and eluate,
multiple myeloma patients who received a nonmye- including anti-C, D, c, e, and “anti-Rh” (reactive with
loablative conditioning regimen. They found that all cells except Rh-null cells). Granulocyte and platelet
patients with ABO-mismatched transplants had pro- antibodies were also present. Efforts to detect residual
blems with engraftment, including graft rejection, recipient cells produced negative results using DNA
PRCA, and persistence of mixed-lineage chimerism. hybridization techniques that are sufficiently sensitive
They suggested that a prospective study to evaluate to detect the presence of chimerism when the minority
the effects of ABO mismatch on engraftment in the population of cells is present at levels of 1%–5%.
nonmyeloablative setting was needed. Therefore, although production of antibody by a small
residual recipient population could not be completely
excluded, the authors concluded that that the findings
AUTOIMMUNE HEMOLYTIC ANEMIA more likely indicated an autoimmune reaction of
AFTER HEMATOPOIETIC STEM CELL donor origin to blood cells and their precursors that
are also of donor origin. Even a very small percentage
TRANSPLANTATION of residual host cells can produce readily detectable
antibodies, however (see pages 489–490).
Another cause of hemolysis after marrow transplanta- Therapy in this case was innovative and produced
tion is autoimmune hemolytic anemia (AIHA). interesting results. The various components of the
Hemolysis is thought to be due to antibodies produced pancytopenia showed different responses to therapy
484 Immune Hemolytic Anemias

in that the hemolysis and the thrombocytopenia eluate, and anti-c was found in the serum. The authors
responded well to immunosuppressive treatment suggested that the antibodies were probably due to an
initially, but the neutropenia did not. Splenectomy autoimmune reaction of the graft against its own
produced only a transient rise in neutrophil counts, products, but they recognized that they could not
but total lymphoid irradiation produced a fall in neu- prove the exact origin of the antibodies because both
trophil antibody levels, with a corresponding amelio- donor and recipient shared the c antigen.
ration of the neutropenia. Sokol and associates,17 in another report, described
Sokal and colleagues130 reported the case of an a 37-year-old male with multiple myeloma who
8-year-old boy with Fanconi’s anemia who received a received a BMT from an HLA-matched unrelated
BMT from an unrelated donor. The patient’s blood donor 8 months after diagnosis. Seven months later,
group was O, ccDEe, and the donor’s was AB, ccddee. the patient developed warm antibody AIHA
Sustained engraftment was achieved, but the patient (WAIHA), which required treatment with pred-
developed grade III GVHD, a virus-associated hemo- nisolone and blood transfusions.
phagocytic syndrome, and immune hemolytic Drobyski and coworkers131 reported on seven adult
anemia. The onset of the immune hemolysis was patients who developed AIHA from a total of 236
6 months after transplantation. The DAT was positive, patients who received T-cell–depleted marrow grafts
broadly reactive RBC antibodies were found in the (Table 12-9 A,B,C). The onset of AIHA was at a median

TABLE 12-9 AUTOIMMUNE HEMOLYTIC ANEMIA AFTER HEMATOPOIETIC CELL TRANSPLANTATION

A. Treatment of Autoimmune Hemolytic Anemia and Clinical Outcome

Clinical Follow-up after Diagnosis of


UPN Treatment Response AIHA (Months) Clinical Outcome

070 Steroids NR 2 Death due to candidal sepsis


123 Steroids/immunoglobulins/EPO CR 57 Off therapy for 3 years
154 Steroids NR
Steroids/splenectomy NR
Steroids/azathioprine NR 84 Off therapy for 4 years
Steroids/accessory splenectomy NR
Steroids/cyclosporine NR
Steroids/EPO CR
178 Steroids NR 0.7 Death due to Gram neg. sepsis
295 Steroids/EPO PR 5 Death due to sepsis, ARDS
Steroids/splenectomy/EPO PR
419 Steroids/immunoglobulin/EPO PR 41 On low-dose steroids
Steroids/splenectomy PR
434 Steroids
Immunoglobulin
Prosorba column
EPO PR 9 Death due to DIC
ATG secondary to AIHA
Danazol/vincristine
Plasmapheresis
Steroids/ATG/cyclophosphamide PR

ARDS = acute respiratory distress syndrome; ATG = antithymocyte globulin, DIC = disseminated intravascular coagulation; EPO = erythropoietin; NR = no response;
CR = complete response; PR = partial response

B. Clinical Characteristics at Time of Diagnosis of AIHA

Type of Onset of AIHA History of GVHD Concurrent Immunosuppressive


UPN Age/Sex Disease Treatment (Months Post-BMT) (Acute/Chronic) Illness(es) Medications

070 36/M CML,AP RM 21 II/extensive CMV retinitis Prednisone


123 19/M Myelodysplasia RPM 25 I/extensive Atypical mycobacterial Prednisone, azathioprine
infection
154 18/F AML, 1 CR RM 12 II/none Transverse myelitis, Prednisone
autoimmune thyroiditis,
CMV infection
178 46/F ALL, 1 REL RM 8 II/none Pseudomonas sepsis Prednisone
295 21/M AML, 1 REL RPM 10 I/limited None Prednisone
419 20/M CML.AP RPM 7 II/limited None Prednisone, cyclosporine
434 27/F CML.AP UNR 10 I/limited None Cyclosporine

1 CR = first complete remission; 1 REL = first relapse; AP = accelerated phase; RM = related match; RPM = related partial match; UNR = unrelated.
Immune Hemolysis Associated with Transplantation 485

TABLE 12-9. AUTOIMMUNE HEMOLYTIC ANEMIA AFTER HEMATOPOIETIC CELL TRANSPLANTATION, continued

C. Laboratory Data at Time of Diagnosis of AIHA

DAT
UPN Hematocrit (%) Polyspecific IgG C3 Serum Antibodies

70 20 4+ 3+ 3+ Warm autoantibody, allo anti-C


123 15 3+ 3+ 3+ Warm autoantibody
154 17 4+ 4+ 0+ Warm autoantibody
178 25 2+ 2+ 0+ Warm autoantibody, allo anti-E
295 22 3+ 4+ 3+ Warm autoantibody
419 13 3+ 3+ 3+ Warm autoantibody
434 28 4+ 0+ 4+ Cold autoantibody, (anti-I), allo anti-E

From Drobyski WR, Potluri J, Sauer D, Gottschall JL: Autoimmune hemolytic anemia following T cell-depleted allogeneic bone marrow transplantation. Bone Marrow
Transplant 1996;17:1093–1099.

of 10 months after transplantation (range 7–25 months) conversion from her pretransplant RBC phenotype of
and occurred in 5% of all patients who survived at A1, Cde/Cde to that of the donor, which was O,
least 6 months. Six patients had a warm reacting cde/cde. Two years after transplantation, a positive
autoantibody, while one patient had a cold-reacting DAT was noted, and shortly thereafter the patient
antibody. developed WAIHA that required transfusion with
In the series reported by Chen and colleagues,132 group O, Rh-negative RBCs. No specificity of the anti-
nine of 293 patients (3.1%) developed AIHA after body was noted after autologous and differential allo-
marrow transplantation. (Tables 12-10 and 12-11). geneic adsorptions. At that time, RBC mixed chimerism
Three of the nine patients had matched unrelated was present, with both group O, Rh-negative and A1,
donors, while the other six had matched sibling Rh-positive red cells demonstrable. She was treated
donors; some of the marrow products were T-cell with immunosuppressive therapy consisting of cortico-
depleted. Four patients developed AIHA with cold steroids, azathioprine, and cyclosporine; she was also
antibodies, whereas five developed AIHA with warm treated with intravenous immunoglobulin (IVIG).
antibodies. Cold-antibody AIHA had an earlier onset Mixed-erythrocyte chimerism persisted, and a compen-
(beginning from 2 to 8 months after transplantation), sated WAIHA continued for about 1 year but then
whereas AIHA associated with warm antibodies worsened as immunosuppression was tapered. Shortly
developed 6 to 18 months after transplantation. In thereafter, after differential alloadsorptions to remove
both series, hemolysis was resistant to treatment and the nonspecific red cell autoantibody, anti-S and anti-
overall prognosis was poor, although no patients died Fyb were detected in her serum. One year later, a
as a direct result of hemolysis but rather from associ- mimicking anti-C was also demonstrated. She re-
ated problems such as sepsis and GVHD. mained on immunosuppressive therapy and under-
Wennerberg and associates133 reported on a patient went splenectomy but continued to have a partially
who was transplanted with a minor ABO-incompatible compensated hemolytic anemia. The authors made the
marrow. By day 180 after transplantation, her red cell interesting observation that RBC antigens that were
antigen phenotype was essentially donor type, with a present on recipient RBCs but absent on donor RBCs

TABLE 12-10. RESULTS OF SEROLOGICAL INVESTIGATION OF NINE PATIENTS WITH AIHA AFTER TRANSPLANTATION

DAT Antibody Specificity Blood Group

Case IgG C3d Serum Eluate Donor Recipient

1 + + NS + anti-e Nonspecific B+ A+
2 + + Nonspecific Nonspecific O+ O+
3 + + NS + anti-D anti-D, E + NS A+ A+
4 + + NS + anti-C, c, e anti-D, e O+ O+
5 + + Nonspecific Nonspecific O+ A+
6 – + anti-I Negative O+ A+
7 – + Nonspecific ND O+ O+
8 – – anti-Pr Negative A– O–
9 – + anti-Pr Negative AB+ AB+

Cases 1–5 were warm type and detected at 37°C. Cases 6–9 were cold type and detected by saline methods at 20°C. Patients 2 and 5 also had an allo anti-E
(i.e., donor is E-neg). All Rh specificities were enhanced by papainized cells. Patient 6 was lytic at 20°C in saline; patient 7 lysed papainized cells at 20°C; patients
8 and 9 showed no lysis.
ND = not done; NS = nonspecific.
From Chen FE, Owen I, Savage D, et al: Late onset haemolysis and red cell autoimmunisation after allogeneic bone marrow transplant. Bone Marrow Transplant
1996;19:491–495.
486 Immune Hemolytic Anemias

TABLE 12-11. CLINICAL COURSE OF THE PATIENTS

Clinical Association

Case RCAI Type CMV GVHD Relapse Treatment Clinical Outcome Peak Bilirubin (mmol/L)

1 Warm + – – Prednisolone Hemolysis resolved 27


Immunoglobulin Alive and well
2 Warm + – + Prednisolone Resistant hemolysis 623
Immunoglobulin CML relapse
Died from pneumonitis
3 Warm – – + Prednisolone Hemolysis resolved after 42
Splenectomy splenectomy
Alive and well
4 Warm – – – Immunoglobulin Hemolysis eventually resolved 30
Azathioprine but required further donor
Splenectomy marrow infusion
Total lymphoid irradiation Alive with no hemolysis
5 Warm – – + Prednisolone Also developed ITP 427
Immunoglobulin Died from multiple thromboembolism
Splenectomy
Vincristine
6 Cold – + – Prednisolone Resistant hemolysis 694
Immunoglobulin Died with GVHD and sepsis
7 Cold + – – Nil Relapsed with lymphoid blast crisis 27
Subsequently reinduced
8 Cold – + – Nil No overt hemolysis 20
Resolved
9 Cold + + + Prednisolone Hemolysis resolved but died from 70
GVHD liver

Note that in patient 5, recipient cells were detected 2 months after hemolysis developed but no relapse of CML. In cases 1, 2, and 3, recipient cells were detected
12.2 and 9 months, respectively, before hemolysis. In patient 9, there was transient cytogenetic relapse at the time of hemolysis, both of which resolved after the
cyclosporine dose was reduced.
From Chen FE, Owen I, Savage D, et al: Late onset haemolysis and red cell autoimmunisation after allogeneic bone marrow transplant. Bone Marrow Transplant
1996;19:491–495.

included D, C, S, and Fyb. If the immune-stimulating Godder and coworkers136 reported that 18 of 40
event was the presence of the minor subpopulation of patients (45%) developed de novo chronic graft-vs.-
recipient RBCs, it is interesting that no anti-D or clearly host disease (cGVHD) after partially mismatched-
distinguishable alloanti-C was detected, especially as related donor BMT. Of those developing cGVHD, five
these are considered more immunogenic than are S and patients (28%) presented with hemolytic anemia on
Fyb. The alternative explanation was that the immuniz- days 112–272 after transplantation (median = 168
ing event was transfusions with group O, Rh-negative days). Four of these patients also had thrombocytope-
RBC, most of which would likely be of the Rh pheno- nia (median platelet count = 50,000/μL). Patients with
type rr (cde/cde) and would thus not carry the D or C hemolytic anemia were HLA-mismatched with their
antigens. Whether the RBC autoantibody was pro- donors for one antigen (n = 1), two antigens (n = 3),
duced by cells of the immune system of the donor or and three antigens (n = 2) and were all sex mis-
recipient was not determined. matched. Signs of hemolysis were present, but few
Pratt and Kinsey134 described an 8-year-old boy serologic findings were reported. Two patients had a
who developed AIHA 6 months after an allogeneic positive DAT, and one of these had a positive IAT. In
bone marrow transplant. He required 4 years of spite of limited evidence of an immune-mediated
immunosuppressive therapy before remission of process, all patients responded to intensive immuno-
hemolysis. suppressive therapy.
Graze and Gale135 reported evidences of auto- Tamura and colleagues137 reported a case of cold
immunity in six patients with chronic GVHD after agglutinin syndrome (CAS) after allogeneic BMT in a
BMT, including the presence of rheumatoid factor and 36-year-old male (Fig. 12-17). The cold agglutinin titers
a positive antinuclear antibody (ANA) test. Four of the recipient and the donor before transplant were
patients developed a positive DAT and clinical evi- less than 4 and 4, respectively. Three weeks after trans-
dence of hemolysis, and the authors suggested that plantation, acrocyanosis developed, and there was evi-
AIHA might occasionally complicate chronic GVHD. dence of hemolysis with a decrease in hemoglobin
Insufficient data were provided to document AIHA, level to 5.3 g/dL, undetectable serum haptoglobin,
however. and an increase in both serum indirect bilirubin and
Immune Hemolysis Associated with Transplantation 487

40 55
mg mg
PSL

512

256
512
256
CA titers

<4

<4

<4

<4
64

16
8
12 RBC
FIGURE 12-17. Clinical course. PSL, predonisolone; CA titers, cold transfusion
agglutinin titers. (From Tamura T, Kanamori H, Yamazaki E, et al: 10

Hb (g/dL)
Cold agglutinin disease following allogeneic bone marrow
transplantation. Bone Marrow Transplant 1994;13:321–323.) 8

4
0 25 50 75 100 125 150 175 200
Days post-BMT

LDH. The DAT was positive with anti-C3, the cold of Evans’s syndrome after umbilical cord stem cell
agglutinin titer was elevated to 512, and the specificity transplantation of an 8-month-old male with X-linked
of the antibody was anti-Pr. Engraftment of donor lymphoproliferative disease. He received multiple
marrow cells was proven by day 22 using cytogenetic courses of IVIG, anti-Rh D immunoglobulin, a pulse
analysis. The patient avoided exposure to cold and of high-dose corticosteroids, and cyclosporine with
was treated with prednisolone. Signs of hemolysis some improvement of the hemolytic anemia, but no
gradually subsided, and on discharge on day 174, the improvement of the thrombocytopenia. Addition of
cold agglutinin titer had decreased to less than 4. vincristine resulted in long-term resolution of throm-
Au and associates138 reported on a 22-year-old bocytopenia and anemia.
woman with CML in second chronic phase who Some cases that have been reported as autoimmune
underwent a marrow transplant from a matched unre- hemolysis11,40 are cases of passenger lymphocyte syn-
lated donor. Eleven months after transplantation, she drome, for which the term “autoimmune” is used
developed an abrupt, painful acrocyanosis up to the inappropriately according to the definitions provided
thighs and elbows. The peripheral blood film showed earlier in this chapter.
extensive RBC agglutination. A cold agglutinin was
present to a titer of 8192 at 4°C, with a thermal ampli-
tude up to 30°C. She was treated with high-dose MIXED CHIMERISM AND ITS
prednisolone, cyclophosphamide, intravenous immu- SIGNIFICANCE IN THE CAUSATION OF
noglobulin, and cessation of cyclosporine. Plasma- RED CELL ANTIBODIES AND IMMUNE
pheresis was unsuccessful because of extensive HEMOLYSIS AFTER HEMATOPOIETIC
extracorporeal agglutination, and exchange transfu-
sion did not reduce the cold agglutinin titer. The CELL TRANSPLANTATION
patient finally died of infection.
Azuma and coworkers139 described a 10-year-old In Greek mythology, Chimera was a monster with a
male with severe aplastic anemia who was treated lion’s head, a goat’s body, and a serpent’s tail
with a marrow transplant from his HLA-matched (Fig. 2-18). In medical terminology, the term chimera is
sister. On post-BMT day 67, a diagnosis of CAS was used to designate an organism whose body contains
made, and he was treated with plasmapheresis on an cell populations derived from different individuals of
emergency basis. Corticosteroids after plasmapheresis the same or different species, occurring spontaneously
had a transient effect. At a second episode of hemo- or produced artificially.146 Thus, all successful allo-
lysis 6 months after BMT, immunosuppressive geneic transplants result in the creation of chimeras.
therapy with cyclophosphamide plus corticosteroid When it was recognized that some allogeneic BMT
was administered successfully without negative effect recipients not only had hematopoietic cells of the donor
on engraftment. but also retained some of their own hematopoietic
Cases of AIHA have been reported after allogeneic system,147,148 there was a need for a term to distinguish
marrow transplantation for aplastic anemia,140 auto- these chimeras from those with complete donor
logous marrow transplantation for recurrent Hodgkin’s hematopoiesis. The terms generally used are mixed
disease,141 a mismatched-related allogeneic PBSC trans- hematopoietic chimera or mixed chimera, in contrast to
plant for chronic myelogenous leukemia,142 a partially complete chimera or donor chimera.146,149-151
matched cord blood stem cell transplant for osteo- Mixed chimerism is detected with increasing fre-
petrosis,143 and T-cell–depleted haploidentical trans- quency as methods for detecting a minor population
plantation for severe combined immunodeficiency.144 of cells become more sensitive. Techniques that have
Diagnoses have included Evans’s syndrome141,142,145 been used include cytogenetic markers, red blood cell
and fatal autoimmune pancytopenia.140 antigens, immunoglobulin allotypes, HLA typing,
Dovat and colleagues145 reported the development persisting RBC antibodies, fluorescence in situ hyrbi-
488 Immune Hemolytic Anemias

hematopoietic stem cell transplantation. Thus, RBC


antibodies can be derived from cells of the immune
system of the donor, the recipient, or both (Table 12-
12). Donor-derived alloantibodies produced as part of
the passenger lymphocyte syndrome are thought to
be produced by lymphocytes that are transfused with
the donor marrow product, proliferate, and produce
antibody. The syndrome resolves after the passenger
lymphocytes (which are not engrafted) reach the ends
of their life spans. Hemolysis has its onset within the
first few weeks after BMT and is transient. In contrast,
other patients develop donor-derived alloantibodies
that are produced by the immune system of the donor
long after transplantation, presumably by engrafted
cells of the donor’s immune system. In still other
patients, residual cells of the transplant recipient’s
FIGURE 12-18. Chimera, the Greek mythologic creature that, immune system produce RBC antibodies.
according to one description, was a lion in front, a goat in the middle,
and a snake in the back. She devoured many men and animals and was
ultimately killed by consent of the gods. In medical science, the term Alloantibodies Produced by Engrafted
chimera is used to designate an organism whose body contains cell Cells of the Donor’s Immune System
populations derived from different individuals of the same or different
species, occurring spontaneously or produced artificially. (From Petz In seven of the 12 patients reported by Ting and asso-
LD: Immunohematologic problems unique to bone marrow ciates29 who developed non-ABO red cell alloantibod-
transplantation. In Garratty G (ed): Red Cell Antigens and Antibodies.
Arlington, VA: American Association of Blood Banks, 1986:195–229.)
ies after BMT, the antibodies were first detected after
days 45–330 and seemingly were produced by cells of
dization, fluorescence-activated cell sorting (FACS) the engrafted marrow.
analysis, in situ hybridization of Y-chromosome,152 Esteve and coworkers154 reported a 16-year-old boy
and numerous molecular biology techniques.150,153 who was group O, Rh-positive (phenotype ccDEe)
Documenting and characterizing donor and recipient and was transplanted with marrow from a group A,
cell populations in the patient after allogeneic BMT Rh-negative donor who had had four pregnancies
was used originally to document engraftment of the and whose serum contained anti-D and anti-C. Seven
donor marrow, but such studies also have the poten- months after transplantation, anti-D was detected in
tial to do the following: his serum, which was well characterized as being of
IgM immunoglobulin class. The antibody persisted
1. Better our understanding of mechanisms of graft for about ten months but disappeared one month
rejection and marrow failure. after cyclosporine was discontinued as GVHD pro-
2. Detect the recurrence of leukemia and evaluate phylaxis. The patient did not develop anti-C, possibly
whether recurrent leukemia occurs in donor or due to the lack of an immunizing stimulus, because
recipient cells. the patient’s RBC did not bear the C antigen. The
3. Determine the clinical significance of mixed authors commented that the long follow-up period
chimerism. without appearance of an IgG component suggests a
4. Provide insights regarding the mechanisms of defect in the immunoglobulin isotype switching
tolerance and GVHD. mechanism.
5. Provide information concerning the kinetics of Heim and colleagues155 reported a 22-year-old man,
engraftment in various disease states or after dif- blood group O, Rh positive (R2r), who received bone
ferent preparative regimens. marrow from his blood group A1, Rh-negative (rr)
6. Determine the origin of cells of the marrow micro-
environment.
TABLE 12-12. SOURCE OF NON-ABO RED CELL
RED CELL ALLOANTIBODIES OTHER THAN ALLOANTIBODIES FOLLOWING HEMATOPOIETIC
ABO PRODUCED BY ENGRAFTED CELLS OF CELL TRANSPLANTATION
THE DONOR’S IMMUNE SYSTEM OR
Donor-Derived
BY RESIDUAL CELLS OF THE PATIENT’S Passenger lymphocyte syndrome
IMMUNE SYSTEM AFTER HEMATOPOIETIC Alloantibodies produced by engrafted cells of the donor’s
STEM CELL TRANSPLANTATION immune system
Recipient-Derived
Alloantibodies produced by residual cells of the patient’s
It is important to appreciate that either complete immune system
chimerism or mixed chimerism can exist after
Immune Hemolysis Associated with Transplantation 489

HLA-identical sister for treatment of acute lympho- antigen, so the antibodies could have been donor-
cytic leukemia. Three months after transplantation, derived antibodies produced by passenger lym-
the patient was found to be a mixed chimera, with phocytes or recipient-derived antibodies produced by
0.5% of the RBCs still of the host’s type. Four months residual cells of the patient’s immune system.
after transplantation, three different Rh antibodies Immunization in these cases was probably related to
(anti-D, -E, and -G) were detected. It was evident that red cell transfusion received after BMT. The source of
the engrafted marrow had produced these antibodies, the antibody was also uncertain in one patient who
and because the patient had received only Rh-nega- had a panagglutinin between days 13 and 32.
tive RBC transfusions, it appears that he had become
immunized to his original red cells. Alloantibodies Produced by Residual
Lasky and associates54 reported four Rh-positive Cells of the Patient’s Immune System
patients who received bone marrow from Rh-negative
donors. Two developed anti-Rh antibodies 12 and Girelli and colleagues157 reported a group O, Rh-
15 months after BMT, respectively. Both patients negative female who received an allogeneic bone
received at least four Rh-positive platelet concentrates marrow transplant from an HLA-compatible group O,
within the first months after BMT. Rh-positive donor (CcDee). No unexpected antibodies
Niethammer and coworkers156 reported a 9-month- were found in the patient’s serum before BMT, and the
old boy who received a BMT from his mother for patient had never been transfused, although she had
severe combined immunodeficiency (SCID). The had two pregnancies with Rh-positive fetuses. On day
mother was group O, Rh negative (ccdee), and the 5 after transplanation, the RBC phenotype of the
patient was group O, Rh positive (CcDee). As is not patient was CcDee due to persistence of RBCs trans-
uncommon after BMT for SCID, incomplete fused in the donor marrow product. From day 8 to day
chimerism developed, with 100% of the red cells 9, the hemoglobin dropped from 9.3 g/dL to 6.1 g/dL,
being of the patient’s type, while chromosome analy- and laboratory evidences of hemolysis were present.
sis revealed 4% XX chromosomes and HLA typing On day 11, anti-D was detected in the patient’s serum,
revealed that the circulating lymphocytes had the the red cell phenotype was now ccddee, and the DAT
HLA type of the mother. Four months after trans- was negative. She was transfused with two units of
plantation, serologic tests revealed anti-D and anti-C Rh-negative blood, and signs of hemolysis gradually
and also a weakly positive reaction by indirect subsided, only to return on day 21 coincident with evi-
antiglobulin test against the mother’s RBCs and all dences of erythropoiesis by the newly engrafted
other Rh-negative RBCs tested. Hemolysis developed marrow. The RBC phenotype was now CcDee, and
as manifested by erythroid hyperplasia of the anti-D could be eluted from the patient’s RBC.
marrow and reticulocytosis without an increase in Hemolysis gradually subsided, and the anti-D became
hematocrit. The anti-D and anti-C were most likely undetectable in the serum on day 35 but was still
produced by the immune system of the donor against detectable on the RBCs until day 46. In this case, the
antigens on the residual RBCs of the patient. It is anti-D could only have been produced by the patient’s
difficult to determine what role, if any, the weak, residual immune system. The antibody apparently
broadly reactive RBC antibody played in the patient’s caused hemolysis of the Rh-positive RBCs that were
clinical course. transfused with the donor marrow product; later, it
Wennerberg and colleagues133 reported a patient caused transient hemolysis of Rh-positive RBCs pro-
who became a mixed chimera after allogeneic BMT duced by the newly engrafted marrow until the anti-
from an unrelated donor. The patient developed AIHA body disappeared. The second episode of hemolysis is
3 years after BMT and, 1 year later, alloantibodies of analogous to that found after some cases of major ABO
donor origin directed against red cell antigens of the blood group incompatibility.
recipient (anti-S and anti-Fyb). Moore and associates158 reported a 17-year-old boy
The distinction between antibody production by who was transplanted with marrow from his HLA-
passenger lymphocytes that do not engraft and anti- identical brother. Both recipient and donor were blood
body production by engrafted cells of the donor type O, Rh positive. The patient’s serum contained
marrow seems clear in many cases on the basis of the anti-Lua on day 23, anti-E on day 28, and anti-K on day
time of onset of the antibody and the duration of its 31 after transplantation. The antibodies became weaker
production. In some instances, however, the distinc- beginning on day 40, and by day 48, all three were
tion is uncertain, and perhaps both can occur in some undetectable. The patient had been transfused with
patients. For example, Hows and associates6 13 units of RBC between 2 and 8 months before trans-
described patients who developed Rh antibodies plantation. Retrospectively, the donors of those units
within the first three weeks after BMT that persisted were typed for RBC antigens. One was positive for K,
for up to one year. and four were positive for E. None was positive for
Ting and coworkers29 reported on three patients in Lua. The patient was also exposed to the E and K anti-
whom alloantibodies were first observed between gens on transfused RBCs on days 11 and 13 after trans-
days 12 and 18 after transplanation, but neither the plantation, respectively. Cytogenetic studies indicated
donor nor the recipient had the corresponding complete marrow engraftment, and RBC phenotyping
490 Immune Hemolytic Anemias

6 months after BMT indicated that the RBCs of donor patients each had one RBC antibody, and one patient
origin were negative for K, E, and Lua antigens. The had two antibodies. The specificities were anti-E (two
authors suggested that the antibodies had been pro- antibodies), anti-Jka, anti-M, and anti-Lu14. The RBC
duced by residual plasma cells of the patient, possibly antibody formation rate was 0.1% per unit of RBCs
as a result of cooperation between elements of residual transfused. The authors did not comment on the
recipient lymphoid cells and cells of the engrafting source of the antibodies. Bar and coworkers53
donor marrow. Gm allotyping did not help in deter- reported that none of 230 patients developed unex-
mining the origin of the antibodies, as both the marrow pected antibodies after BMT.
donor and the recipient had the same allotypes.
In some instances, residual cells of the patient’s
immune system are presumably responsible for the OTHER IMMUNOHEMATOLOGIC
production of antibodies against antigens on donor ABNORMALITIES AFTER
RBCs, even though very sensitive techniques fail to HEMATOPOIETIC STEM CELL
detect the residual host cells. In the series reported by TRANSPLANTATION
Petz and coworkers,151 RBC antibodies persisting
more than 6 months after BMT served as the basis of
diagnosing mixed chimerism in seven patients. In two Evidences of Autoimmunity
of these patients, immunoglobulin allotyping also after Hematopoietic Stem Cell
indicated mixed chimerism, whereas in the other five Transplantation
patients, immunoglobulin allotyping either was unin-
formative or was not performed. Anti-A or anti-B anti- Immune system imbalance can occur during the
bodies were present in six patients and caused a reestablishment of immune function and the hema-
positive DAT without hemolysis in two patients, a topoietic system after hematopoietic stem cell allo-
positive DAT with transient hemolytic anemia in one grafting. The development of humoral and/or
patient, and a delayed onset of erythropoiesis in three cell-mediated immunity directed against smooth
patients. muscle, epidermis, mitochondria, cell nuclei, cardio-
Similar findings were reported by Izumi and col- lipin, liver-kidney microsomes, human leukocyte anti-
leagues,159 who described a 26-year-old male who had gens, thyroid antigens, cytoskeletal proteins, and
preexisting alloantibodies to E and c. He received a other cellular components has been reported.9,164-166
BMT from a donor whose Rh phenotype was E+, c+. Rouquette-Gally and colleagues167 studied 53 long-
From about one month after the transplant onward, term survivors of allogeneic BMT, 40 of whom had
he showed mild hemolysis due to the antibodies. The chronic GVHD. They found a high frequency of various
patient-derived antibodies remained detectable by autoantibodies, especially considering the young age of
both DAT and IAT even 20 months after BMT, even their patients (mean age = 20.3 years; range, 4–33). The
though immunomagnetically isolated peripheral cir- prevalence of antinuclear, anti–smooth muscle, antimi-
culating B cells were 100% of donor origin. The patient tochondria, anti–liver-kidney microsome, and anti-
received prednisolone from day 221 onward, and epidermal antibodies was 62.2%, 49%, 11.3%, 5.6%,
thereafter, the signs of hemolysis disappeared. and 11.3%, respectively. The screening for anti-DNA,
von Tol and associates160 reported that IgG of reci- anti-extractable nuclear antigen, anticentromere, and
pient origin persisted in 15 of 18 informative recipi- anti-salivary gland duct antibodies was negative. The
ents (83%) until last follow-up (i.e., for several years presence or absence of acute GVHD made no difference
after BMT) despite the fact that the circulating B cells in the frequency of autoantibodies. The authors pointed
appeared to be entirely of donor origin at that time. out that although there are clinical features of GVHD
Other investigators have reported similar results.161,162 that mimic collagen vascular disease, the biological
Thus, even in this era of sensitive molecular techno- autoimmune profile of GVHD is different.
logy, persistence of RBC antibodies and immunoglo-
bulin allotyping, when applicable, appear to be Immune Thrombocytopenia and
among the most sensitive techniques for indicating Leukopenia after Hematopoietic
the existence of residual cells of the recipient’s
immune system.
Cell Transplantation
There are numerous reports of immune cytopenias
Incidence of Red Cell Antibody Formation after hematopoietic stem cell transplantation, and in
after Hematopoietic Cell Transplantation some of these, the origin (i.e., donor or recipient) of
the antibodies has been determined or reasonably
The incidence of RBC antibody production after BMT inferred. As defined at the beginning of this chapter,
appears to be low. Abou-Elella163 reported that only 4 antibodies of donor origin against antigens on donor
of 193 patients (2.1%) developed RBC alloantibodies cells, or antibodies of recipient origin against antigens
from the date of admission for BMT until the date of on the recipient’s cells, may be considered autoanti-
hospital discharge (48.5 ± 14.9 days for autologous bodies. In contrast, antibodies produced by cells of
and 58.7 ± 25.9 days for allogeneic transplants). Three host origin against antigens on donor cells should be
Immune Hemolysis Associated with Transplantation 491

considered alloantibodies, as should antibodies of Graft-vs.-Host Disease


donor origin that react with antigens on the recipient’s and Post-transplant Immune
cells. One must keep in mind the difficulty in deter- Thrombocytopenia
mining the origin of the antibodies, especially as
many of the case reports were published before sensi- There have been reports that immune thrombocytope-
tive techniques were described for detecting the per- nia after BMT could be associated with GVHD. In this
sistence of host cells after transplantation. setting, autoantibodies rather than alloantibodies
have been implicated.
First and coworkers177 reviewed cases of isolated
Autoimmune Thrombocytopenia thrombocytopenia after BMT in 65 fully engrafted
Minchinton and associates168 described a patient who patients surviving at least 60 days after transplana-
received an allogeneic BMT from a donor who had an tion. Nine patients had transient thrombocytopenia
IgM antibody that reacted with his own platelets. The with recovery by day 90, whereas 15 patients had
patient had no platelet autoantibodies before grafting, chronic thrombocytopenia, in which a normal platelet
but after transplantation, an IgM platelet antibody count was not achieved at any time during the first
was found in the patient’s serum that reacted with her 4 months after transplantation. The chronic syndrome
circulating platelets and with platelets from the bone carried a high mortality and had a high association
marrow donor. After BMT, the peripheral blood neu- with both severe (grades 3 to 4) acute GVHD and
trophil count was more than 1 × 109/L by the 23rd day, chronic GVHD. In most cases, bone marrow biopsies
but severe thrombocytopenia persisted until comple- demonstrated adequate numbers of platelet precur-
tion of a course of treatment with IVIG, which was sors, suggesting peripheral platelet destruction or
started on day 87. It seems reasonable to infer that the ineffective thrombopoiesis. Platelet antibodies were
donor’s autoantibody-producing cells were engrafted found in four of eight thrombocytopenic patients
by the patient, resulting in a true autoimmune throm- tested (one of two transient, three of six chronic). Six
bocytopenia (donor vs. donor). patients underwent platelet survival studies (two
Minchinton and Waters169 subsequently studied 14 transient, four chronic), and all showed a markedly
patients who received autologous buffy coat or bone decreased platelet half-life (mean of 6 hours compared
marrow grafts and 32 patients who received allogeneic with normal platelet survival of 6.5 days).
bone marrow grafts. There was a high incidence (52%) Anasetti and colleagues178 studied platelet and
of antibodies to circulating platelets in the early post- fibrinogen kinetics and antiplatelet antibodies in 20
graft period. The presence of postgraft antibodies did patients between 60 and 649 days (median = 90 days)
not predict the development of a cytopenia, however, after transplantation (19 allogeneic transplants and one
evidently because of variable in vivo activity of the anti- syngeneic transplant). Seventeen patients had isolated
body and the ability of the engrafted bone marrow to thrombocytopenia (<100,000/μL). Platelet survival
compensate for antibody-mediated cell destruc- studies indicated that the major mechanism for throm-
tion.170,171 Antibodies demonstrated after autografting bocytopenia was increased platelet destruction. Platelet
were, by definition, autoantibodies. Antibodies in the antibodies bound to the patients’ platelets were present
allografted patients were shown, by immunoglobulin in five of the 12 patients studied. Patients with platelet
allotyping, to be of marrow donor type and on this basis antibodies had lower platelet counts and shorter
were thought to be autoantibodies.169 platelet survivals than patients without platelet anti-
Bierling and colleagues172 described a patient who bodies. Platelet-bound autoantibodies were present in
developed immune thrombocytopenia after allo- five of six patients with grade II–IV acute or chronic
geneic BMT. The patient’s serum contained an IgG GVHD but were not present in six patients free of
antibody that reacted with the patient’s platelets GVHD. The authors concluded that persistent throm-
(donor origin) and with both of the patient’s HLA- bocytopenia after BMT is most often secondary to
identical siblings. The authors suggested that this was increased platelet destruction mediated by multiple
an autoimmune thrombocytopenia, apparently con- mechanisms and that platelet autoantibodies are found
cluding that the platelet antibody was produced by in patients with acute or chronic GVHD.
engrafted donor cells and reacted with platelets of
donor origin. Alloimmune Thrombocytopenia—
Benda and associates173 reported on a patient who Recipient vs. Donor
developed autoimmune thrombocytopenia after an
allogeneic BMT from her HLA-identical sister. An IgG Bierling and associates179 described a patient who had
autoantibody was found that bound to the platelet mild, clinically asymptomatic immune thrombocy-
glycoprotein IIb/IIIa, similar to reactions in patients topenia after allogeneic BMT for CML. A number of
with idiopathic autoimmune thrombocytopenia. In studies suggested that all hematopoietic cells were of
addition, the authors detected an autoantibody to donor origin. These included cytogenetic studies and
HLA class I proteins. molecular analysis of peripheral blood cells, purified
Other investigators have also reported cases of B- and T-lymphocyte subpopulations, and bone
autoimmune thrombocytopenia after BMT.9,174-176 marrow colonies. Nevertheless, the authors identified
492 Immune Hemolytic Anemias

anti-HPA-5b (anti-Bra) of recipient origin that recog- body against the neutrophil-specific antigen NB1 as
nized donor platelets. The antibody remained well as additional neutrophil antibodies of uncertain
detectable more than 3 years after BMT. Further specificity present in lower titer. Despite treatment
studies using adsorption-elution techniques demon- with granulocyte-macrophage colony-stimulating
strated a small amount of recipient RBCs, and the factor, the patient remained neutropenic and died of
CML chimeric transcript was detected by the poly- polymicrobial sepsis and aspergillosis 38 days after
merase chain reaction until day 867 after transplanta- transplantation. The authors suggested that the
tion, indicating persistent mixed chimerism of low patient probably became alloimmunized as a result of
level without hematologic relapse. Thus, this case of granulocyte transfusions but also pointed out that
immune thrombocytopenia was clearly shown to be a patients with large granular lymphocytes have a high
result of a recipient-vs.-donor reaction rather than incidence of autoantibodies, including neutrophil
being an autoimmune thrombocytopenia. antibodies. Neutrophils from the marrow donor
Panzer and coworkers180 described a patient who expressed the NB1 antigen, but the authors were
developed immune thrombocytopenia more than one unable to phenotype the patient’s neutrophils to
year after successful allografting for CML. Serologi- determine whether the anti-NB1 was an autoantibody
cally, the antibody was found to be directed against or an alloantibody. In contrast to the poor outcome in
the donor platelet antigen PlA1. Determining the phe- this case, Warkentin and coworkers186 reported the
notypes of the platelets of the patient and his family successful engraftment (without delayed neutrophil
indicated heterozygosity (PlA1/A2) in the parents and, recovery after transplantation) of an NA1-positive
accordingly, a 25% probability that the patient inher- bone marrow in a patient with aplastic anemia who
ited both PlA2 haplotypes. Further, residual cells of had anti-NA1.
host origin were found in the patient’s excised spleen. Tosi and colleagues187 described a 26-year-old male
These findings suggested that this was a recipient- with chronic myelogenous leukemia who was the
derived immune reaction against the donor’s PlA1 recipient of an HLA-compatible marrow from a
platelet antigen rather than an autoimmune reaction. matched unrelated donor. Cyclosporine and a short
Szymanski and colleagues181 also described a course of methotrexate were administered as GVHD
patient who had anti-HPA-5b before BMT. It persisted prophylaxis. Good platelet engraftment was obser-
after BMT, indicating that the patient’s antibody- ved, but the WBC count remained extremely low,
producing cells had survived the severe conditioning ranging from 0.2 to 0.4 × 109/L. On day 33 after trans-
regimen. plantation, surface-bound neutrophil antibody was
detected by immunofluorescence, although the
patient’s serum was negative when tested with
Immune Neutropenia random neutrophils. A repeat test 6 days later was
Immune neutropenia after BMT has been described by still positive, although with decreased intensity. The
a number of investigators.9,128,171,182,183 Antigranulocyte granulocytopenia resolved without additional
antibodies can occur in the post-transplant setting and, immunosuppressive therapy by day 42, at which time
although often benign, can be the cause of otherwise the test for neutrophil antibody was negative. The
unexplained neutropenia after autologous or allogeneic case represents an example of single-lineage anti-
BMT.171 Minchinton and Waters169 reported that body-mediated cytopenia, as neither RBCs nor
immunoglobulin allotyping on allografted patients platelets were affected and their engraftment
indicated that the antibodies were of marrow donor occurred normally.
type and were therefore autoantibodies.
Koeppler and Goldman184 reported on a 32-year-old
male who developed isolated neutropenia six months THE DIFFERENTIAL DIAGNOSIS OF
after allogeneic BMT for CML from his HLA-matched HEMOLYSIS AFTER HEMATOPOIETIC
brother. The presence of granulocyte-specific IgM and
IgG antibodies in the patient’s serum indicated an STEM CELL TRANSPLANTATION
immune-mediated basis for the neutropenia. The
patient was in continuous complete remission, sug- A note of caution is warranted concerning the diagno-
gesting that the antibody was an autoantibody pro- sis and management of immune hemolysis caused by
duced by the donor’s immune cells. Immunoglobulin ABO antibodies after BMT. Hemolysis occurring in
allotyping was not performed to document the source the various characteristic clinical settings should
of the antibody-producing cells, however. quickly lead to a tentative diagnosis, which is then
Stroncek and associates185 described a 13-year-old readily confirmed by simple serologic tests. The major
girl with large granular lymphocytosis and chronic pitfalls are that these hemolytic syndromes are rather
neutropenia who was treated with granulocyte trans- unusual and often begin abruptly. Accordingly, their
fusions before undergoing a transplant with bone sudden and unexpected onset sometimes results in a
marrow from a partially matched, unrelated donor. delay in diagnosis, while more common problems—
After the transplant, the patient remained neu- such as GVHD and veno-occlusive disease of the
tropenic, and her serum contained a high-titer anti- liver—are inappropriately considered as reasons for
Immune Hemolysis Associated with Transplantation 493

elevated bilirubin and anemia. The delay in institut- system. Also, because at least several days are required
ing appropriate therapy to minimize hemolysis can for proliferation of the transfused lymphocytes, hemo-
result in unnecessary morbidity, particularly renal lysis during the first few days after BMT is not caused
insufficiency (Table 12-13). by the passenger lymphocyte syndrome.
The passenger lymphocyte syndrome caused by
Minor ABO Blood Group Incompatibility antibodies of blood group systems other than ABO is
rare after hematopoietic stem cell transplantation,
DIAGNOSIS OF PASSENGER LYMPHOCYTE although somewhat more common after solid organ
SYNDROME transplantation (see page 502).
Laboratory Tests. The causative antibody is usually
Clinical Setting. The diagnosis of passenger lym- detectable in the patient’s serum and in a RBC eluate
phocyte syndrome is usually not difficult. Indeed the throughout the period of time when hemolysis is
diagnosis generally should be anticipated because of occurring. One point that deserves emphasis, however,
the characteristic clinical setting. That is, patients are in is that signs of hemolysis (e.g., a fall in hemoglobin
the early post-transplant period after a hematopoietic and hematocrit, elevated bilirubin, and elevated LDH)
stem cell transplant in which there is a minor ABO may occur for a short time (from 1 day to as long as 3
blood group mismatch between donor and recipient. days) before the responsible antibody becomes
Additional risk factors are the use of cyclosporine detectable (see pages 461 and 470). Accordingly, post-
without methotrexate for post-transplant GVHD pro- transplant follow-up of patients transplanted with a
phylaxis, the use of stem cells obtained from peripheral minor mismatched hematopoietic stem cell product
blood, and a nonmyeloablative conditioning regimen. should include both laboratory tests to detect hemoly-
Hemolysis characteristically occurs between days 5 sis and serologic studies to detect RBC antibodies.
and 20 after transplantation. When the onset of hemo- Antibody in the Donor Marrow Product. Lasky
lysis is delayed beyond several weeks after transplan- and associates54 reported on 13 patients who received
tation, it appears more likely that antibody is being a marrow transplant from a donor with a minor ABO
produced by cells of the donor immune system that are incompatibility. One patient was managed with
engrafted or by residual cells of the patient’s immune exchange transfusion before BMT using RBCs of the
donor’s type to prevent hemolysis caused by iso-
hemagglutinins in the marrow product. Five patients
TABLE 12-13. THE DIFFERENTIAL DIAGNOSIS were managed by centrifuging the bone marrow to
OF HEMOLYSIS FOLLOWING HEMATOPOIETIC remove plasma and thus reduce the amount of
STEM CELL TRANSPLANTATION antibody. Two of the seven patients who received
uncentrifuged bone marrow experienced minimal
Minor ABO blood group incompatibility hemolysis on the first day after transplantation (indi-
Passenger lymphocyte syndrome rect bilirubin 0.9–1.1 mg/dL). One other patient who
Antibody in the donor marrow product received centrifuged marrow developed hemolysis on
Major ABO blood group incompatibility
Immediate hemolysis (lysis of RBCs in donor stem cell product) the sixth post-transplant day, but the delayed onset of
Early post-transplant hemolysis (lysis of RBC in donor stem cell hemolysis in this case suggests that it was due to the
product caused by rebound of isohemagglutinins after passenger lymphocyte syndrome rather than to
transplantation in patients treated with plasma exchange passive transfer of antibody.1
prior to transplantation)
Delayed hemolysis (lysis of RBCs produced by the newly
engrafted marrow caused by residual isohemagglutinins) Major Blood Group Incompatibility
Red cell alloantibodies of blood groups other than ABO
Alloantibodies produced by engrafted cells of the donor’s IMMEDIATE HEMOLYSIS
immune system
Alloantibodies produced by residual cells of the patient’s Hemolysis at the time of a marrow transplant can, of
immune system
Autoimmune hemolytic anemia
course, be expected unless the marrow product is
Passive transfer of antibody depleted of RBCs and/or the serum antibody is
Plasma transfusion removed from the patient by plasma exchange. Even
Platelet transfusion with rather small volumes of residual RBCs, some
Intravenous immunoglobulin signs of hemolysis can occur, but this does not result
Other causes of passive transfer of antibody; packed RBCs,
intravenous anti-D, and antilymphocyte globulin in serious morbidity.
Drug-induced hemolytic anemia
Microangiopathic hemolytic anemia
Miscellaneous additional causes of hemolysis following bone DELAYED HEMOLYSIS
marrow transplantation
Infusion of cryopreserved stem cell products In patients who are treated with pretransplant plasma
Infusion of dimethylsulphoxide (DMSO) and free plasma exchange and transplanted with a marrow that has not
hemoglobin been depleted of RBCs, a rebound of the antibody can
Clostridium perfringens septicemia
Hemolysis associated with hemodialysis
occur after transplantation causing hemolysis of the
RBCs that were transfused with the marrow. Even more
494 Immune Hemolytic Anemias

serious hemolysis due to rebound of hemagglutinins causing hemolysis. Because platelet transfusions and
has been reported in patients who also were transfused IVIG commonly are administered to BMT patients in
with ABO-incompatible RBCs before BMT to absorb the post-transplant period, however, hemolysis from
isohemagglutinins. Such hemolytic episodes usually passive transfer of antibody can occur during the
occur between days 3 and 10 after transplantation. same time period as the passenger lymphocyte syn-
Another cause of delayed hemolysis, beginning on drome. Indeed, Kim and coworkers188 described such
day 35 to 105, is most likely a result of destruction of a patient, who received a minor ABO-incompatible
RBCs being produced by the newly engrafted marrow BMT and was treated with IVIG in the early post-
by persistence of isohemagglutinins. transplant period.
Hemolysis after Platelet Transfusion. Passive
transfer of antibody can occur as a result of transfu-
LABORATORY TESTS sion of minor ABO-incompatible platelets, as each
Laboratory tests indicative of hemolysis (see Chapter platelet concentrate obtained from a unit of whole
2) alert one to the diagnosis, which is confirmed blood contains about 50–70 mL of plasma, and
readily by the presence of a positive DAT and the plateletpheresis products can contain about 350 mL of
expected isohemagglutinins in the patient’s serum plasma. The use of incompatible platelets is some-
and in an eluate from the patient’s RBCs. times unavoidable, such as when when inventories
are inadequate or when HLA or platelet-specific anti-
bodies are causing refractoriness and the only HLA-
Red Cell Alloantibodies of Blood Groups matched or cross-matched–compatible platelet donors
Other Than ABO are ABO incompatible.189 Cases of severe hemolysis
after transfusion of platelets with ABO-incompatible
Although ABO blood group antibodies are the most plasma have been reported.189-201
important cause of hemolysis in the post-transplant Hemolysis occurs when the donors have high titers
period, hemolysis also can be caused by alloantibo- of antibodies against the recipient’s ABO blood group
dies of other blood group systems. These episodes of antigens. For example, Pierce and colleagues193
hemolysis occur in less well-defined clinical settings reported two instances of severe immune hemolysis
and could be caused by donor-derived or recipient- after transfusion of ABO-incompatible platelet con-
derived antibodies, as described previously. centrates. One of the reactions resulted in dissemi-
nated intravascular coagulation and proved fatal,
Autoimmune Hemolytic Anemia after while the other resulted in hemolysis of an estimated
Hematopoietic Cell Transplantation 40% of the patient’s RBC mass. They found that the
IgG anti-A titer of one donor was 32,000 and the anti-B
Autoimmune hemolytic anemia can occur after titer of the other donor was 16,000.
hematopoietic stem cell transplantation and is unre- In the case reported by Reis and Coovadia,195 the
lated to the presence or absence of blood group plateletpheresis donor’s plasma had an anti-B titer of
incompatibility between the donor and recipient. This 4096 by indirect antiglobulin testing. In the patient
topic is discussed earlier in the chapter, and further described by Conway and Scott,192 the donor’s saline
information regarding the diagnosis of AIHA is pre- anti-A titer was 8192, and the titer of dithiothreitol-
sented in Chapters 2, 5, and 6. treated serum was 4096. Transfusion of a platelet-
pheresis product with a volume of 200 mL resulted in
severe hemolysis, disseminated intravascular coagu-
Passive Transfer of Antibody lation, and renal failure requiring dialysis.
Human plasma and immunoglobulin derivatives The patient reported on by Murphy and associ-
contain alloantibodies to human blood group antigens ates189 received two transfusions of plateletpheresis
that can be acquired passively by patients who receive products 25 days apart from the same donor. Stored
these products. Most commonly of concern in the serum from the donor, taken at the time of the first
setting of marrow transplantation are antibodies that transfusion, had an anti-A titer of 512 by saline agglu-
might be infused as part of the donor marrow product, tination and a titer of 2048 by indirect antiglobulin
platelet transfusions, or IVIG therapy. These antibodies test. The patient received 155 mL of plasma with the
affect cross-matching, antibody screening, and anti- first platelet product and 448 mL with the second.
body identification testing in addition to potentially Hemolysis occurred after both transfusions, and after
causing hemolysis. the second transfusion, the patient’s hemoglobin
The antibodies that are usually responsible for dropped from 11.4 g/dL to 6.0 g/dL in 1 day. She
hemolysis are anti-A, anti-B, and anti-D, which are developed renal failure, although dialysis was not
also the antibodies most likely to cause passenger required and her renal function returned to normal.
lymphocyte syndrome. Frequently, the clinical setting McLeod and coworkers191 reported on a particularly
is valuable in determining the cause of hemolysis interesting patient who developed hemolysis after
because symptoms and signs occur immediately after receiving only four platelet concentrates, each contain-
the passive transfer of antibodies that are capable of ing about 50 mL of plasma. The patient developed
Immune Hemolysis Associated with Transplantation 495

serious hemolysis with hemoglobinuria, a drop in Although hemolytic reactions to ABO-incompatible


hemoglobin from 14.0 g/dL to 8.0 g/dL, and renal platelets are apparently uncommon, they can be
insufficiency with a rise in creatinine from 1.6 mg/dL serious and even fatal, physicians should therefore be
to 6.0 mg/dL. Two of the platelet donors were found by aware of this possibility whenever transfusion of such
indirect antiglobulin test to have anti-A to a titer of platelets is necessary.
10,240. Immune Hemolysis after Intravenous Immuno-
McManigal and Sims196 described a group AB patient globulin. IVIG is commonly administered to patients
who was transfused with 3380 mL of ABO-incompatible after allogeneic BMT, as CMV prophylaxis in seropos-
platelet products during a 4-day period. After transfu- itive patients and as nonspecific antimicrobial pro-
sion of a group O unit of apheresis platelets, she devel- phylaxis. In addition, the immunomodulatory effect
oped classic signs of an intravascular hemolytic of immunoglobulins on GVHD is the subject of clini-
transfusion reaction. Although the volume of RBC cal trials and follows the empiric observation that
hemolyzed was apparently small because the hema- higher immunoglobulin levels are associated with a
tocrit did not change, the manifestations of the transfu- decreased rate of GVHD.203
sion reaction included chills, flank pain, dyspnea, Positive antiglobulin tests have frequently been
hemoglobinuria, and life-threatening tachycardia. The reported with the use of IVIG, but usually signs of
authors contacted the United States Food and Drug hemolysis have been absent or minimal.204-207 A
Administration (FDA) and learned that five deaths number of reports of significant hemolysis after IVIG
caused by ABO-incompatible platelet products had administration exist, however.
been reported in a 4-year period beginning in 1994. Patients who have developed hemolysis generally
Incidence of Hemolysis after ABO-Incompatible have received rather high doses of IVIG. A common
Platelet Transfusions. In spite of the foregoing case dose for treatment of patients with immune cytopenias
reports, one must keep in mind that hemolysis after is 400 mg/kg daily for 5 days.208 A female patient
ABO-incompatible platelet transfusion is not described by Copelan and coworkers208 was treated
common. It could become a more common problem as with 400 mg/kg daily for 10 days; after the sixth day,
the use of apheresis platelets increases, although this the DAT became positive, an eluate revealed anti-D,
has not been documented.199 Indeed, Shanwell and and laboratory evidences of hemolysis were present.
colleagues202 reported that BMT recipients at their During the 10 days of IVIG therapy and the 4 subse-
institution routinely received single-donor platelet- quent days, the patient required 6 units of RBCs to
pheresis products from blood group O donors con- maintain her hemoglobin level above 9 g/dL. Another
taining about 350 mL of plasma. Over a period of 5 patient received IVIG at a dose of 500 mg/kg for 6
years, about 1000 plateletpheresis products with ABO- days. The DAT became positive with anti-IgG on the
incompatible plasma were transfused. Although the fourth day of therapy, and the eluate showed anti-D
researchers noted positive DATs due to passively and anti-A1. Hemolysis developed on the sixth day and
adsorbed anti-A and/or anti-B from donor plasma, could have been aggravated by the subsequent trans-
they found no cases of hemolysis. They studied 11 fusion of a single-donor group O, Rh-positive platelet
group A patients in detail and found that nine devel- concentrate. Indirect antiglobulin titrations on the IVIG
oped a positive DAT and that anti-A could be eluted preparations used revealed anti-A (titer 32), anti-B
from their red cells. None developed hemolysis, (titers 4 to 8), and anti-D (titers 2 to 4).
however. Also, patients of groups A, B, or AB (n = 34) Thomas and colleagues209 reported a case in which a
required no more RBC or platelet transfusions than group A1 patient received 1 g/kg of IVIG on two con-
did patients who were group O (n = 47). secutive days. The following day, signs of hemolysis
More recently, Mair and Benson200 reported on a developed, including the presence of free hemoglobin
group A patient who experienced acute hemolysis, in his serum and urine. His hemoglobin level dropped
with a drop in hemoglobin from 8.4 g/dL to 5.8 g/dL from 15 g/dL to 8.7 g/dL, the DAT became positive, an
in 24 hours after a platelet transfusion from a group O eluate showed anti-A1, and the IVIG batch used con-
donor with a saline anti-A titer of 128. As a result of tained anti-A to a titer of 32 against A1 red cells.
this reaction, they reviewed their experience and Nicholls and associates210 reported on a group A,
stated that the index case was the only identified Rh-positive patient who developed signs of hemolysis
episode of hemolysis that occurred after 46,176 after the fourth course of IVIG, each course consisting
platelet transfusions over a 10-year period (October of 400 mg/kg/day for 5 days. Hemolysis recurred
1986 to October 1986). Given that 21% of all platelets during a fifth course of IVIG and was attributed to
transfused at their medical center are plasma incom- passively transferred anti-A and anti-D. A second
patible, the incidence of identified episodes of hemo- patient also developed hemolysis attributed to pas-
lysis was only one per 9000 incompatible transfusions. sively acquired anti-D and an alloanti-E.
Larsson and associates199 reported a hemolytic reac- Robertson and coworkers207 found that 49% of 47
tion, which was the first that had been recognized at patients who were treated with IVIG after BMT had a
their institution even though more than 6600 single- positive DAT, and 25.5% had a positive IAT.
donor platelet products had been transfused over the Antibodies identified in the serums and eluates were
previous 9 years. anti-A, -B, -D, and -K. Twenty-one lots of IVIG were
496 Immune Hemolytic Anemias

tested, and all contained anti-A and anti-B; two also production.217 The reportability of this case empha-
contained anti-D, and two others contained anti-K. sizes the extreme rarity of hemolysis after transfusion
The observed patterns of anti-D and anti-K reactivity of minor ABO-incompatible RBC.218 Although this
were identical in patients’ sera, in red cell eluates, and possibility must be kept in mind, it is among the least
in the IVIG lots studied. The authors reported “slight likely causes of hemolysis in transplant recipients.
hemolysis” in some patients, but no patient devel- The administration of intravenous anti-D has
oped clinically significant hemolysis. proven to be effective in some Rh-positive patients
Kim and colleagues188 described two BMT reci- with idiopathic thrombocytopenic purpura (ITP).
pients who developed hemolysis after IVIG treatment. Transient and slight signs of hemolysis are found fre-
One of these patients was particularly interesting in quently, but overt clinical hemolysis has been consi-
that he had received a transplant from a minor ABO- dered to be unusual.220,221 A review by Gaines,222
incompatible donor 9 days previous to the IVIG however, described 15 cases of hemoglobinemia
administration. The patient had shaking chills during and/or hemoglobinuria that occurred between
the IVIG administration; hemoglobinuria occurred September 1995 and March 1999. For the 12 patients
5 hours later and persisted for 4 days. Although IVIG for whom symptoms were reported, two did not expe-
probably precipitated the hemolysis, the passenger rience any symptoms, while ten had classic findings
lymphocyte syndrome could have been a contributing associated with acute hemolytic transfusion reactions.
factor. Information about the duration of hemoglobinuria
Other cases in which overt hemolysis occurred after was available for eight patients. Hemoglobinuria per-
IVIG administration have been reported by Nakamura sisted 1 day or less in two patients, 2 days or longer in
and associates,211 Okubo and coworkers,212 Brox and three patients, 3 days or longer in one patient, 7 days
colleagues,213 and Nakagawa and associates.214 or longer in two patients, and 20 days or longer in one
A number of authors have suggested that steps be patient. Between May 1999 and October 1999, sub-
taken to minimize the possibility of hemolysis as a sequent to the time period of the review, the FDA
result of IVIG administration. Several investigators received an additional 26 anti-D IVIG adverse event
have suggested that standards should be set for reports of possible or probable hemoglobinemia
maximum titers of RBC antibodies in IVIG prepa- and/or hemoglobinuria. Three sets of frequency esti-
rations204,215 and/or that a minor cross-match be mates for the development of this complication were
performed using the IVIG preparation to be adminis- calculated (Table 12-14).
tered.209,210 Such standards have not been set, Another potential cause of hemolysis is passive
however, and currently available literature indicates transfer of antibody via antilymphocyte globulin,
that ABO antibodies13,188,208,209,216 and other RBC anti- which is used as an immunosuppressant.17
bodies13,208,216 are found commonly in IVIG. Thus,
cross-matching would seem to be of limited value, as
RBC antibodies would be detected frequently,
Drug-Induced Hemolytic Anemia
although commonly at titers that are benign. Immune reactions to drugs must be considered as a
Determining the titer of red cell alloantibodies in each possible cause when transplant patients develop
lot of IVIG would provide more definitive informa- hemolysis. This topic is reviewed in Chapter 8.
tion, although it would be difficult to determine what
titers should be considered dangerous. Nevertheless,
with further experience, it might be possible to
Microangiopathic Hemolytic Anemia
develop guidelines based on titer and dose. The term microangiopathic hemolytic anemia was first
Because experience has indicated that standard used by Brain and coworkers223 to describe the
doses of IVIG rarely cause immune hemolysis, deter- hemolytic anemia associated with fragmented red
minations of red cell antibody titers in IVIG are not per- blood cells that occurred in patients with microvascular
formed routinely. Physicians should be aware that disease and that often was associated with thrombocy-
hemolysis is a possible outcome, especially with the topenia. These authors suggested that the characteristic
administration of unusually high doses. Other adverse RBC morphology, the accelerated RBC destruction, and
effects of high-dose IVIG are described in Chapter 11. the thrombocytopenia were secondary consequences of
Other Causes of Passive Transfer of Antibody: fibrinoid necrosis and hyaline occlusion of arterioles
Packed RBC, Intravenous Anti-D, and Antilympho- and capillaries in various disease states. Subsequent
cyte Globulin. Inwood and Zuliani55 reported a case experimental work and clinical observations have led
of intravascular hemolysis after transfusion of packed to the acceptance of these concepts.224-227
group O RBCs to a group A patient. The hemoglobin Microangiopathic hemolytic anemia can occur
dropped from 6.5 g/dL to 4.4 g/dL, but the patient in association with a large number of clinical
made an uneventful recovery. The donor’s anti-A1 conditions:228–233
titer was 8192, and subsequently, it was determined
that the donor had participated in a plasmapheresis • Disseminated intravascular coagulation
program in which donors were stimulated for the • Acute renal failure
production of ABO antibodies for subsequent reagent • Radiation nephritis
Immune Hemolysis Associated with Transplantation 497

TABLE 12-14. ESTIMATED FREQUENCY OF HEMOGLOBINEMIA AND/OR HEMOGLOBINURIA


FOLLOWING ADMINISTRATION OF ANTI-D IGIV FOR ITP

Reported Total Estimated Estimated


Source(s) of Data Patients (n) Patients (n) Incidence Rate (%) Reporting Rate (%) Estimated Incidence

Clinical studies 0 528 0.0 NA 0 in 528


Clinical trial 2 137 1.5 NA 1 in 69
FDA*/IMS 13 14,500* NA 0.1 1 in 1115

Reported patients experienced hemoglobinemia and/or hemoglobinuria following anti-D IGIV administration during a specified time period. Total
patients were treated or estimated to have been treated with anti-D IGIV during a specified time period. NA indicates not applicable.
* Data on file, Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, FDA, Rockville, MD:
March 1995–December 1998.
From Gaines AR: Acute onset hemoglobinemia and/or hemoglobinuria and sequelae following Rh(o)(D) immune globulin intravenous administration in
immune thrombocytopenic purpura patients. Blood 2000;95:2523–2529.

• Malignant hypertension bocytopenia, and renal failure. In recent years, to


• Infections with viral or nonviral agents accelerate the initiation of treatment since the advent
• Exposure to a variety of antibiotics of plasma exchange, the diagnosis of TTP-HUS is
• Severe preeclampsia or eclampsia made on the basis of only the principal features—
• Abruptio placenta microangiopathic hemolytic anemia and thrombocy-
• Adenocarcinomas and other malignant tumors topenia (Table 12-15).243
• Hemangiomas HUS-TTP has been reported to occur after total
• Immunologic disorders such as SLE, acute glome- body irradiation, therapy with a number of chemo-
rulonephritis, polyarteritis nodosa, Wegener’s therapeutic and immunosuppressive drugs, and
granulomatosis, and scleroderma among patients who have received a bone marrow or
solid organ transplant.
Frequently, the microangiopathic hemolysis in these
disorders has occurrred in the context of syndromes
DIFFICULTIES IN DIAGNOSIS IN THE
known as the hemolytic uremic syndrome (HUS) or
POST-TRANSPLANT SETTING
thrombotic thrombocytopenic purpura (TTP). In spite
of the imposing list of underlying disorders, these It often can be difficult to establish a diagnosis of
syndromes most frequently occur de novo. transplant-associated TTP (TA-TTP) in hematopoietic
Hemolysis associated with fragmented RBCs cell transplant patients.122,242 These patients fre-
occurs in patients with cardiovascular abnormali- quently have multiple potential etiologies for renal
ties234–237 and as a result of prosthetic cardiovascular dysfunction, fever, and thrombocytopenia, and might
materials.237,238 Because there is no microvascular display only subtle neurological abnormalities. In
disease, however, these disorders are more appropri- addition, the presentation of TA-TTP is highly hetero-
ately called mechanical, traumatic, or fragmentation geneous, ranging from asymptomatic, low-level RBC
hemolysis.238,239 fragmentation to fulminant disease. The diagnosis of
TA-TTP is made most reliably by examination of the
THROMBOTIC THROMBOCYTOPENIC PURPURA peripheral blood film for RBC fragments. Daly and
AND THE HEMOLYTIC-UREMIC SYNDROME coworkers122 suggest that an increase in the number of
AFTER TRANSPLANTATION RBC fragments and the biochemical evidence of
hemolysis over several observations can make the
The importance of TTP and HUS in transplantation is diagnosis more convincing than finding features com-
suggested by the fact that there have been multiple patible with the diagnosis on a single occasion. For
detailed reviews of this topic.122,240-242 this reason, it is essential that blood films be reviewed
TTP and HUS were described initially as distinct and reported by experienced staff and that reliable
disorders. At present, some243 but not all244 investiga- quantitative evaluations or RBC fragmentation be
tors consider TTP and HUS to be different expressions available.
of the same disease process. The precise distinction Zomas and colleagues249 found that mild RBC
between the two syndromes remains somewhat arbi- fragmentation was a common morphologic finding
trary and controversial.240,245,246 after transplantation but that none of their 58 allograft
The classic pentad of TTP consists of fever, or 32 autograft patients developed full-blown throm-
microangiopathic hemolytic anemia, thrombocy- botic microangiopathy. Indeed, Allford and associ-
topenic purpura, renal insufficiency, and neurologic ates242 point out that nearly all patients receiving
findings that often fluctuate in both nature and sever- cyclosoporine have some evidence of microangio-
ity.229,247,248 On the other hand, findings in HUS pathic hemolysis, but the majority do not develop
consist of microangiopathic hemolytic anemia, throm- TTP. Furthermore, these authors have observed
498 Immune Hemolytic Anemias

TABLE 12-15. EVOLUTION OF DIAGNOSTIC CRITERIA FOR TTP

Percentage of Patients

Clinical Sign 1925–1964 1964–1980 1982–1989

Microangiopathic hemolytic anemia 96 98 100


Thrombocytopenia 96 98 100
Neurologic symptoms 92 84 63
Renal disease 88 76 59
Fever 98 59 24

From George JN, El-Harake M: Thrombocytopenia due to enhanced platelet destructon by nonimmunologic
mechanisms. In Beutler E, Lichtman MA, Coller BS, Kipps TJ (eds): Williams Hematology. New York: McGraw-Hill
Health Professions, 1995:1290–1315.

patients with severe, rapidly fatal post-transplant TTP highly significant association of microangiopathy
who had no evidence of hemolysis at the onset of the with severity of acute GVHD and the use of
illness. One group found that LDH was twice the cyclosporine. Galli and associates271 also implicated
laboratory normal in all 22 patients in their series, so cyclosporine administration as a cause of HUS in a
this might be a valuable criterion.250 heart transplant recipient. Hemolysis subsided after
treatment with plasma exchange, but rechallenge with
INCIDENCE OF TA-TTP AFTER HEMATOPIETIC cyclosporine caused recurrence of the microangio-
STEM CELL TRANSPLANTATION pathic hemolysis. Tacrolimus (FK506), an alternative
immunosuppressive drug that is used in marrow and
The exact incidence is difficult to determine due to dif- solid organ transplantation, has also been reported to
ferences in case definition, reporting bias, and missed be associated with HUS and TTP.272,273
diagnoses. Some reported incidence rates have been Paquette and coworkers263 reviewed clinical data
0.28%251 and 4.8%.250 Other studies support an inci- from seven patients diagnosed with severe thrombotic
dence of 5–15%.252,253 microangiopathy and from 409 patients who under-
Rabinowe and coworkers254 reported a series of 168 went BMT during the same time period and survived
adult patients with hematologic malignancies who for at least 100 days afterwards. Univariate analysis
underwent autologous or allogeneic bone marrow revealed an increased risk of thrombotic microangiopa-
transplantation and were investigated for the subse- thy with the use of an unrelated bone marrow donor
quent development of HUS. All patients were condi- (P = 0.02), but no significant association with patient
tioned with cyclophosphamide and total body age or gender, diagnosis, amount of prior chemother-
irradiation. Sixteen patients (9.5%) developed clinical apy, transplant conditioning regimen, or severity of
and laboratory evidence of HUS between three and GVHD. A multivariate exact logistic regression analysis
11 months after BMT. Both hemolytic anemia and revealed that only the type of GVHD prophylaxis had
thrombocytopenia ultimately resolved, but at 18 a significant impact on the risk.
months after diagnosis, some patients had residual Other reports of HUS after BMT indicated that the
creatinine elevations and persistent hypertension. patients were heterogeneous with respect to underly-
Other cases of HUS and TTP have been reported in ing diagnosis, type of BMT (allogeneic or autologous),
both children and adults after allogeneic or auto- pretransplant conditioning regimen, presence or
logous marrow or peripheral blood stem cell absence of GVHD, and use of cyclosporine.274 Allford
transplantation.254-266 and colleagues242 concluded that the role of cyclo-
sporine in the etiology of TTP remains unclear.
RISK FACTORS FOR HUS AND TTP Marshall and Sweny275 reported three cases of HUS
after BMT in which none of the patients was treated
Risk factors for the development of HUS or TTP after with cyclosporine, and Chavers and associates276
BMT have been analyzed. Irradiation of the kidney reported a case of HUS in a renal transplant recipient
(which occurs during total body irradiation) is likely who had none of the factors known to contribute to
to be involved in the pathogenesis.267 In fact, the clin- the development of the syndrome. The etiology is
ical picture of HUS can be indistinguishable from that likely to be multifactorial.277,278
of radiation nephritis.268-270 Holler and colleagues253 Other reports suggest that patients with TA-TTP are
reported intravascular hemolysis with RBC fragmen- more likely to be female and are significantly older than
tation and de novo thrombocytopenia in 49 of 66 allo- patients who do not develop the syndrome. Also,
geneic marrow graft recipients receiving cyclosporine grades II–IV acute GVHD and hepatic veno-occlusive
but none in 11 patients treated with methotrexate for disease are associated with an increased risk of TA-
prophylaxis of GVHD. Risk factor analysis revealed a TTP.122
Immune Hemolysis Associated with Transplantation 499

HUS and TTP have been reported after Manifestations of radiation nephritis develop bet-
renal,276,279–281 liver,253,272,282,283 and heart271 transplan- ween 6 and 12 months after irradiation.267
tation. Schwarz and coworkers281 reviewed the clini- The time of onset of HUS or TTP after organ trans-
cal course of 700 patients with renal transplants and plantation is variable, occurring within the first 1 to 3
concluded that cyclosporine treatment did not weeks after transplantation in some patients271,279,280,296
increase the incidence of HUS. and 3 to 16 months after transplantation in
A number of chemotherapeutic agents have been others.272,276,283
reported to cause HUS or TTP. The most commonly
implicated drug is mitomycin, but these syndromes Miscellaneous Additional Causes of
have also been reported in association with carbo-
platin,284 gemcitabine,284 bleomycin, cisplatin,
Hemolysis after Hematopoietic Stem
vinca-alkaloids, FK-506, daunorubicin, and methyl- Cell Transplantation
CCNU.272,273,285-287 INFUSION OF CRYOPRESERVED BONE MARROW
Ticlopidine use has been implicated in numerous OR PERIPHERAL BLOOD STEM CELL PRODUCTS;
cases of TTP.288,289 Quinine sensitivity is a relatively INFUSION OF DIMETHYLSULPHOXIDE (DMSO)
newly recognized cause of HUS and is unique in that AND FREE PLASMA HEMOGLOBIN
an immune mechanism has been implicated.290-294
Although high concentrations of DMSO cause hemo-
MANAGEMENT AND PROGNOSIS OF TA-TTP lysis in vitro and in vivo,297-299 concentrations used in
the modern practice of marrow and peripheral blood
There is no consensus on what constitutes appropriate stem cell transplantation usually produce only mild
therapy for patients with TA-TTP.122,242 Almost all clini- side effects. These include nausea, chills, fever, hyper-
cians stop cyclosporine or tacrolimus, however, and tension, bradycardia, shortness of breath, and tran-
platelet transfusions are avoided if at all possible. Many sient substernal chest tightness. Contaminating RBCs
centers employ plasma exchange, although this strategy that are lysed during the cryopreservation procedure,
is based neither on data nor on logical therapy, as no however, are an obvious cause of hemoglobinemia
substance has been identified in the plasma that and hemoglobinuria immediately after administering
appears to be associated with disease pathogenesis.242 the stem cell product.
Response rates to plasma exchange have been con- Smith and colleagues300 reviewed 33 consecutive
siderably lower than in classical TTP, and long-term patients who had received unfractionated cryopre-
survival rates have been disappointing. Overall, just served autologous bone marrow. Gross hemoglobin-
under half of patients in nine reported case series have uria was noted in all 33 patients, three of whom
achieved a complete or partial remission, but fewer developed acute renal failure with renal histopatho-
than half of patients who respond to plasma exchange logy typical of an acute hemolytic transfusion reac-
experience prolonged survival.122 Many of the tion. Although previous investigators had attributed
patients in these reports died of causes other than TA- nephrotoxicity in patients receiving autologous bone
TTP—most commonly, infection, GVHD, and multi- marrow to hemolysis caused by DMSO, Smith and
system organ failure syndrome. associates implicated the hemolysate content of the
cryopreserved marrow. Subsequently, they adopted a
DIFFERENTIATING HUS/TTP FROM IMMUNE procedure to remove RBCs from the marrow harvest
HEMOLYSIS IN THE POST-TRANSPLANT SETTING specimens.
Burger and coworkers301 reported acute hemoglo-
HUS and TTP are readily distinguished from immune binemia and hemoglobinuria after infusion of autolo-
hemolytic syndromes after transplantation. Major gous buffy coat products that contained RBCs and
points of differentiation are a lack of red cell antibod- were cryopreserved using DMSO. Although the recip-
ies in HUS or TTP (with the exception of quinine- ient’s free plasma hemoglobin levels after infusion of
dependent antibodies in quinine-induced HUS) and the products were as high as 932 mg/mL, no overt
the presence of fragmented RBCs in the peripheral renal problems resulted. The products contained ele-
blood film. vated levels of free hemoglobin due to the presence of
The time of onset is often helpful in making a dif- lysed RBCs. Accordingly, to minimize RBC contami-
ferential diagnosis, as most immune hemolytic syn- nation, the authors recommended a policy of
dromes occur earlier after hematopoietic stem cell mononuclear cell separation for patients undergoing
transplantation than do HUS and TTP, which typically autologous marrow transplantation.
develop at a median time of 5 months after transplan- Kessinger and colleagues302 evaluated 100 consecu-
tation (range 3 to 7 months).295 Other reports indicate tive autologous peripheral blood stem cell trans-
the median time of onset as 163 days274 or 61 days.253 plants and determined that hemoglobinuria occurred
Only occasionally do cases occur within the first in 92%, elevated serum bilirubin in 43%, and elevated
month after BMT.274,277 serum creatinine in 15% of patients. Larger volumes
HUS caused by mitomycin has its onset at a median of transplanted cells with concomitantly larger doses
time of 1 year from the beginning of treatment.285 of DMSO and lysed RBCs were both strongly related
500 Immune Hemolytic Anemias

to a greater number of side effects of the marrow infu- Additional causes are thermal red cell injury such as
sion (P < 0.0001). The authors tested a number of pro- might be caused by overheated dialysate, osmotic red
cedures to decrease the volume transfused and the cell injury caused by hypotonic dialysate, and partial
number of lysed RBCs in the infusate. All methods obstruction within the extracorporeal circuit.
eliminated renal toxicity, but several resulted in
delayed engraftment. A method involving a repeat cen-
trifugal apheresis of the collected product, harvesting IMMUNE HEMOLYSIS ASSOCIATED
the entire buffy coat and the first 60 mL of discharged WITH SOLID ORGAN
cells that have a visible RBC content, eliminated renal TRANSPLANTATION
toxicity and allowed timely engraftment.
Passenger Lymphocyte Syndrome
CLOSTRIDIUM PERFRINGENS SEPTICEMIA
The passenger lymphocyte syndrome, with clinical
Clostridium perfringens is a recognized but rare cause and laboratory findings very similar to those found
of septicemia and remarkably severe intravascular after BMT, has been reported on numerous occasions
hemolysis303-307 and is more likely to occur among after transplantation of kidney,24,311–324 liver,325–330
immunocompromised patients. Ifthikaruddin and lung,331–334 heart,335–337 heart-lung,334,338,339 spleen,332
associates308 reported on a 54-year-old female with pancreas,312,340 and pancreas-spleen341 (Table 12-
acute myeloblastic leukemia who underwent an 16).24,311-324 Ramsey, in a review in 1991,44 found that
autologous BMT but failed to engraft. She was fol- among minor ABO-incompatible transplants, 61% of
lowed as an outpatient and was dependent on blood cases involved group O donors and group A recipi-
product support. Approximately 4 months after BMT, ents, 22% percent involved group O donors and group
she was admitted to the hospital with cellulitis of the B recipients, and 17% percent involved group AB
left inner thigh. On admission, her hemoglobin was patients receiving non-AB organs.
10.6 g/dL. Antibiotics were promptly prescribed, but Hemolytic anemia after organ transplantation is
the patient had signs of septic shock and became more frequently encountered in proportion to the lym-
unresponsive within hours, during which time hemo- phoid mass transplanted.45,342 Ramsey44 found that the
lysis of her total red cell mass occurred. Just 5 hours frequencies of antibodies and hemolysis were lowest in
after admission, the hemoglobin was 2.3 g/dL, but kidney transplant patients (17% and 9%, respectively),
this value was attributed to free hemoglobin in the intermediate in liver transplant patients (40% and 29%,
plasma because the peripheral blood film failed to respectively), and highest in heart-lung transplant
show any intact RBCs. The patient also developed dis- patients (both, 70%). Borka and coworkers24 reported
seminated intravascular coagulation and expired on that hemolysis attributed to the passenger lymphocyte
the day of admission. Such massive hemolysis with syndrome developed in 22 of 237 of their patients (9%)
destruction of essentially all circulating red blood cells who received a minor incompatible renal transplant.
strongly suggests a diagnosis of Clostridium perfrin- Salerno and colleagues334 reported that in heart-lung
gens septicemia, and this diagnosis was confirmed the transplantation, the incidence of hemolysis from
following day by the results of blood cultures. donor-derived anti-ABO antibodies is as high as 70%.
Chaplin303 reviewed the causes of massive intravas- Triulzi and associates327 reported that five of nine
cular hemolysis and discussed a patient with “total patients (56%) receiving an ABO minor-mismatched
intravascular hemolysis” caused by Clostridial perfrin- liver transplant developed donor-derived antibody and
gens infection. He reviewed six cases in which the hemolysis. It is remarkable that the few lymphocytes
hematocrits were reported to be 0%, 0%, 0.6%, 1%, less transplanted with some donor organs are able to prolif-
than 2%, and less than 5%, respectively. The diagnosis erate sufficiently to produce adequate quantities of anti-
of Clostridial sepsis should be considered at once body to cause hemolysis of the recipient’s RBCs within
when gross hemoglobinemia and hemoglobinuria a week or two after the transplantation. Antibody can
and a rapid fall in hemoglobin level are found, as the be produced even when the donor organs have been
immediate institution of appropriate antibiotics and perfused copiously with preservative solution and, just
aggressive supportive care could be life saving. before transplantation, with saline.325,343,344

HEMOLYSIS ASSOCIATED WITH HEMODIALYSIS IMMUNOGLOBULIN ALLOTYPES


OF ANTIBODIES CAUSING THE PASSENGER
Occasionally, hemodialysis is indicated for patients LYMPHOCYTE SYNDROME
who have received a solid organ or marrow transplant.
Hemolysis in chronic renal failure is persistent but The origin of cells producing immunoglobulins can be
usually mild.309 Aggravation of hemolysis can occur in investigated by analysis of polymorphic epitopes
the hemodialysed patient for a number of reasons, (allotypes) on the immunoglobulins in instances in
however. Acute intravascular hemolysis has been which the donor and recipient express different phe-
caused by an indwelling hemodialysis catheter.310 notypes.160 Several groups have confirmed that the
TABLE 12-16. ABO ANTIBODIES FROM SOLID ORGAN TRANSPLANTS: TOTAL REPORTED CASES AND COMBINED FREQUENCIES
OF ANTIBODY (Ab) AND HEMOLYSIS (H) IN STUDIED SERIES OF ABO-UNMATCHED ORGANS

Donor-Recipient Kidney Liver Heart-Lung Heart


ABO Blood Other Total
Groups Cases %Ab %H Cases %Ab %H Cases %Ab %H Cases %Ab %H Cases* Cases

Total 46 17 9 45 40 29 7 70 70 2 40 11 6 106
(33)† 24/144‡ 15/165§ (35) 36/90 33/115§ (7) 7/10 7/10 (1) 2/5 1/9§ (3) (79)
O-A 22 23 8 26 50 44 6 75 75 2 50 14 3 59
(15) 8/35 4/51 (24) 18/36 16/36 (6) 6/8 6/8 (1) 2/4 1/7 (2) (48)
O-B 10 9 5 8 40 25 0 0 0 0 0 0 3 21
(9) 2/22 1/22 (5) 8/20 5/20 — 0/1 0/1 — 0/1 0/1 (1) (15)
O-AB 2 50 50 4 50 0 1 100 100 — — 7
(1) 1/2 1/2 (2) 1/2 0/2 (1) 1/1 1/1 (4)
A-AB 1 6 5 3 50 17 — 0 — 0 — 4
(1) 1/16 1/20 (2) 3/6 2/12 0/1 (3)
B-AB 3 10 0 2 33 0 — — — 5
(2) 1/10 0/11 (0) 2/6 0/6 (2)
Not given 8 2 — — — 10
(5) (2) (7)

* Three pancreas (O-to-B, 1H), two spleen (O-to-A, 2H), and one lung (O-to-A) transplant.
† Number with hemolysis from among those reported; the kidney cases included one kidney-pancreas graft with hemolysis.
‡ Number with Ab or H/total number studied in all series; in the kidney and liver transplants, the total numbers of cases evaluated for the frequencies of Ab and H are greater than the sum of the ABO rows,
because several series did not specify ABO groups in all cases.
§ Some patients were evaluated for H but not for Ab; the kidney series included two kidney-pancreas grafts without hemolysis.
From Ramsey G: Red cell antibodies arising from solid organ transplants. Transfusion 1991;31:76–86.

501
502 Immune Hemolytic Anemias

antibodies causing passenger lymphocyte syndrome Seltsam and colleagues349 reported the occurrence of
were produced by donor lymphocytes and not by RBC alloimmunization in two of four patients who
those of the recipient.44,323,343-346 received different organs from an immunized donor.
The donor was a 58-year-old woman who was group
SEROLOGIC FINDINGS O, D+, K-, and Fy(a-). Her serum contained anti-K and
anti-Fya, and several of her organs were transplanted
One should be aware that hemolysis is usually abrupt to different patients. The recipient of her liver devel-
in onset and that serologic abnormalities might not be oped anti-K, and anti-Fya was eluted from her RBCs. A
evident before the onset of hemolysis. Ramsey44 patient who received a pancreas-kidney graft also had
reported that a positive DAT, serum antibodies, and RBCs sensitized with anti-Fya. The latter patient devel-
hemolysis have generally been found at the same oped mild hemolysis.
time. In our experience, we have been rather disap-
pointed to find that performing DATs and serum anti- POSSIBLE FACTORS AFFECTING HEMOLYSIS
body studies on patients in the postoperative period
are not helpful in predicting the onset of hemolysis. Patient gender and age did not affect the likelihood of
Also, as with hematopoietic stem cell transplantation, either antibody appearance or hemolysis after minor
hemolysis precedes the ability to detect the relevant ABO-incompatible organ transplants.44 Patients of
antibodies for a day or so in some cases. Triulzi and blood groups A1 and A2 had similar frequencies of
coworkers327 reported that hemolysis preceded the hemolysis, contrary to the expectation that in group A2
first positive DAT in four of five liver transplant recip- recipients, the reduced expression of RBC antigen
ients who developed passenger lymphocyte syn- might protect against hemolysis by antibodies from
drome (donors group O, patients group A) by a mean group O organs. The patients’ secretor status was also
of 1.8 days (range, 0 to 4 days). Similarly, Borka and evaluated, as patients who are genetic ABH secretors
colleagues24 reported that antibodies are found after have more ABH antigen in their plasma than nonse-
hemolysis has been present for “several days.” cretors, and the suggestion has been made that soluble
Accordingly, the most informative laboratory findings ABH antigen could serve to neutralize circulating anti-
are a sudden drop in hemoglobin and hematocrit body. This could not be proven on the basis of limited
without evidence of a significant source of bleeding. available data, however.
These findings are associated with an elevation of the Among patients reviewed by Ramsey,44 those
serum bilirubin and LDH. The DAT and serum anti- receiving cyclosporine did not have a significantly
body tests, if not immediately positive, will become so higher frequency of antibodies (30%) than those
during the ensuing days. receiving azathioprine (17%). The frequency of hemo-
The onset of hemolysis is generally between 3 and lysis in patients taking cyclosporine, however, was
24 days, regardless of whether the antibodies are higher (17% vs. 3%). Subsequently, Povlsen and asso-
in the ABO or Rh blood group systems. The findings of ciates315 reported two cases of severe but self-limited
the DAT in 43 patients with minor ABO-incompatible hemolysis among 34 kidney transplant patients
transplants revealed that 36 patients had IgG on their treated with cyclosporine but no cases among 108
RBCs, 35 had complement, and 28 had both.44 patients treated with azathioprine. Borka and cowork-
In three kidney and six liver transplant patients, graft ers24 reported that 91% of their 22 renal transplant
anti-A was solely of A1 specificity. Three of these patients suffering from the passenger lymphocyte syn-
instances occurred in A1 patients. The other six occurred drome were treated with cyclosporine, in contrast to
in A2 or A2B patients. Because hemolysis caused by anti- their entire incompatible group of 237 patients, in
A1 is rare,347 a number of authors have suggested that whom only 65% were treated with this agent.
anti-A could have been absorbed onto RBCs and tissues, Mazarra and colleagues340 reported on a patient
with anti-A1 left circulating as a separate antibody.44,338 who developed anti-B without hemolysis from day 11
Alternatively, Brecher and associates347 postulated that to day 31 after a pancreas transplant, and Lundgren
anti-A1 seen after organ transplantation could be the and associates312 described a patient who developed
result of cyclosporine-induced altered T-lymphocyte fulminating hemolysis caused by donor-derived anti-
regulation of B cells leading to proliferation of hereto- A after a renal allograft. Neither patient received
fore rarely detected clones of clinically significant anti- cyclosporine.
A1–producing B cells. Tacrolimus (FK-506) has also been associated with
Hemolysis caused by Rh antibodies has more com- donor-derived antibodies and hemolysis in liver trans-
monly been reported after solid organ transplantation plant recipients. Bradley and coworkers350 reported that
than after BMT. Specificity has most often been anti- the DAT was positive in three of five liver transplant
D,44,317,328,339 but anti-c316 and anti-e44,348 also have patients who had the test performed postoperatively.
been noted. RBC eluates contained anti-A or anti-B donor-derived
Hareuveni and coworkers329 reported clinically RBC antibodies. The donor-derived antibodies appeared
significant hemolysis associated with the passenger a mean of 14 days after transplantation, and one patient
lymphocyte syndrome after liver transplantation developed mild hemolysis requiring a transfusion of 2
caused by anti-Jka. units of group O RBCs.
Immune Hemolysis Associated with Transplantation 503

CLINICAL FEATURES Forty-eight hours before collection of the kidney, the


donor had been transfused with E-positive RBCs.
Hemolysis is characteristically acute in onset, and Immunosuppressive therapy, which was started on the
although many cases are mild and self-limited, RBC day of transplantation, consisted of cyclosporine and
transfusion is often required. Ramsey44 reported that prednisone. In the third post-transplant month, the
the median number of units transfused in 18 renal patient was admitted to the hospital because of severe
transplant patients was 6.5 (range of 1 to 18). In seven hemolytic anemia with a hemoglobin of 2.6 g/dL, a
liver transplant patients, the number of units trans- total bilirubin level of 2.2 mg/dL, and a lactate dehy-
fused ranged from 2 to 11 units, and four heart-lung drogenase level of 1039 IU/L. Serum haptoglobin was
patients received 16–24 units. In some instances, more below 10 mg/dL, and creatinine was 2.3 mg/dL.
severe hemolysis has occurred with resultant renal The patient’s DAT was positive, and anti-E was
failure requiring dialysis.282,312,333,346,351 detected in the serum and in an eluate from his RBCs.
In contrast to BMT, wherein the patient’s RBCs are Cyclosporine dosage was reduced, high-dose pred-
replaced by those produced by the donor marrow, nisone therapy was started, and he was transfused
incompatible red cells continue to be produced by the with 7 units of group A, E-negative RBCs. Hemolysis
solid organ transplant recipient. Nevertheless, hemo- gradually resolved. Three months later, the DAT was
lysis is generally short-lived, evidently because the still positive and anti-E was still present in the
lymphocytes transferred with the donor organ are patient’s serum, but there were no signs of hemolysis.
able to proliferate only temporarily and are not Twelve months after transplantation, the DAT was
engrafted permanently. In kidney transplant patients, negative and anti-E could not be detected.
the final positive DATs were seen 2 to 13 weeks after Au and colleagues330 described a 45-year-old male
operation (median, 5), and the last reactive serum (group A1, Rh-positive) who received an orthotopic
specimens were detected at 3 to 23 weeks after trans- liver transplant from a male cadaveric donor (group
plantation (median, 5.5). In liver transplant patients, O, Rh-positive). Six units of group A RBCs were trans-
the reactive DAT or serum was last detected 10–50 fused in the peritransplant period. On postoperative
days after surgery (median, 20).44 day 6, the hemoglobin level was normal, but on day 8,
the patient developed shock and jaundice accompa-
UNUSUAL CASES nied by a dramatic fall in hemoglobin to 3 g/dL. The
patient was transfused with 8 units of group O RBCs.
Some particularly interesting cases are worth noting. The peripheral blood showed numerous spherocytes,
Jacobs and colleagues352 reported on the simultaneous the DAT was strongly positive, and anti-A was
occurrence of the passenger lymphocyte syndrome present to a titer of 512. The donor’s titer of anti-A was
and a delayed hemolytic transfusion reaction 16 days found to be 2560. Hemolysis persisted for 3 weeks,
after transplantation of an ABO minor mismatched and 4 more units of group O RBCs were given, after
liver. Serologic studies identified a positive DAT and a which signs of hemolysis gradually resolved.
donor-derived anti-A (i.e., passenger lymphocyte syn-
drome). In addition, a recipient-derived anti-E was
identified in the serum and eluate. DONOR-DERIVED ANTIBODIES PRODUCED LONG
Bracey and Van Buren353 reported anti-A of donor AFTER TRANSPLANTATION
lymphocyte origin in three group A recipients of
Larrea and associates355 reported a case in which
organs from the same group O donor. The recipient of
donor-derived anti-E and immune hemolysis was
the donor’s liver developed hemolysis caused by anti-
not detected until the third month after trans-
A 10 days after transplantation, and two recipients of
plantation. Accordingly, the antibody was likely
the same donor’s kidneys developed hemolysis due
produced by cells of the donor that had resulted in
to anti-A on days 11 and 13, respectively. A similar
microchimerism (see the earlier discussion in this
case was reported by Ramsey and associates,317 who
chapter).
reported anti-D in two Rh-positive patients receiving
Swanson and coworkers348 reported one patient who
renal grafts from an Rh-immunized donor. One
had severe hemolytic anemia of 4 months’ duration
patient developed overt hemolysis, whereas evidence
caused by anti-e. Ramsey and colleagues317 docu-
of hemolysis was minimal in the other patient.
mented the presence of anti-D in two Rh-positive
Bapat354 reported a patient who had the passenger
patients for 6 months after each had received a renal
lymphocyte syndrome caused by anti-A after renal
transplant from the same Rh-sensitized cadaver donor.
transplantation and in whom thrombocytopenia
occurred coincident with the onset of brisk hemolysis
and persisted throughout the period of ongoing MANAGEMENT OF THE PASSENGER
hemolysis. Platelet-specific antibodies were not LYMPHOCYTE SYNDROME
detected.
The patient was a 30-year-old male, group A, D pos- Anticipation of the passenger lymphocyte syndrome
itive (Cde/cDE), who received a kidney transplant is important. Unfortunately, the titer of antibody in
from a group A, D-positive, E-negative cadaver donor. the donor is not reliable as a means of prediction of
504 Immune Hemolytic Anemias

the occurrence of antibody or hemolysis in the post- and in the postoperative period in minor ABO-
operative period.327,338 incompatible transplants, especially if the donor is
Appropriately selected blood products must be group O and the patient group A.327 We have not used
used throughout in all organ transplants, as indicated this option because we do not find a high incidence of
in Table 12-17.356 Red cell components for transfusion patients with severe hemolysis, and we try to con-
must be ABO identical or compatible with recipient serve our group O RBCs because they might be in
serum, regardless of the organ donor’s type. When relatively short supply.
donor and recipient are of different ABO blood Washed RBC are generally unnecessary because the
groups, however, special attention is given to the small amount of plasma in packed RBCs is not likely
selection of plasma and platelet components. These to represent a significant source of antibody.327
should be ABO compatible with both recipient RBCs Hemodialysis has been necessary for acute renal
and donor organ tissue cells to avoid transfusing anti- failure caused by hemolysis.333 Death due to dissemi-
body that might contribute to RBC hemolysis. nated intravascular coagulation that develops during
Cryoprecipitate can be given without regard to ABO hemolysis beginning nine days after a renal transplant
type because this product contains only a small has been reported.357,358
volume of plasma.
Because most cases of the passenger lymphocyte
syndrome are the result of a minor ABO blood group OTHER IMMUNOHEMATOLOGIC
incompatibility between donor and recipient, it is ABNORMALITIES AFTER SOLID ORGAN
wise to monitor such patients between approximately TRANSPLANTATION
days 3 and 15 after transplantation, as in marrow
transplants (see Table 12-4).
Hemolysis can generally be managed by transfu- Immune Hemolytic Anemia Caused by
sion of group O RBCs, avoidance of ABO-incompati- Multiple Alloantibodies Produced by
ble plasma products, and maintenance of adequate Donor Lymphocytes Masquerading as
renal perfusion. Corticosteroids are often used empir- Autoimmune Hemolytic Anemia
ically. In patients with more severe hemolysis, plasma
exchange may be used to decrease antibody titer.44,327 Ishikura and coworkers359 studied a patient who
Also, RBC exchange may be used to decrease the received an ABO-matched liver transplant at age
volume of circulating incompatible RBCs.327 Jenkins 11 months because of congenital biliary atresia. He
and associates351 have recommended that, if more received multiple transfusions during the surgery and
than 2 units of RBC need to be transfused within the postoperative course. On postoperative day 198,
first 24 hours after the onset of hemolysis, RBC massive gastrointestinal bleeding occurred, and the
exchange should be considered. Subsequent transfu- DAT was positive for the first time (33 days after the
sions should be of group O RBC until antirecipient last blood transfusion of RBCs). The antibody eluted
antibody has disappeared. from the patient’s RBCs showed panagglutination,
Other potential approaches may be considered. including panel cells, cord cells, Rh-null, and -D-RBCs.
When the surgery to perform the transplant is likely to Two days after transfusion of least incompatible
utilize large volumes of RBC, one may consider using RBCs, severe hemolysis occurred with progressive
group O RBCs for transfusion during the procedure anemia (hemoglobin 4.0 g/dL), hyperbilirubinemia

TABLE 12-17. BLOOD SELECTION FOR RECIPIENTS OF ABO-MISMATCHED ORGANS

Recipient ABO Donor ABO Red Blood Cells Fresh-Frozen Plasma No. 1 Platelets* No. 2 Platelets†

O A O A,AB B,O
B O B,AB A,O
AB O AB A,B,O
A O A,O A,AB B,O
B A,O AB B,A,O
AB A,O AB A,B,O
B O B,O B,AB A,O
A B,O AB A,B,O
AB B,O AB B,A,O
AB O AB,A,B,O AB A,B,O
A AB,A AB A,B,O
B AB,B AB B,A,O

* No. 1 = Preferred ABO choice for platelets.


† No. 2 = Alternative ABO choice for platelets, in order of preference, if preferred ABO is not available.
From Petz LD, Calhoun L: Preparation for blood and blood product replacement. In Busuttil RW, Klintmalm GB (ed): Transplantation of the Liver.
Philadelphia: WB Saunders, 1996:442–448.
Immune Hemolysis Associated with Transplantation 505

(direct, 16.6 mg/dL; indirect, 6.2 mg/dL), hemoglo- tive kidney grafts have produced anti-D, apparently as
binuria, and high LDH (2546 IU/L). The hemolysis an anamnestic response to the residual red cells in the
subsided after therapy with plasmapheresis, whole graft.369-372 Ramsey and coworkers373 reported that
blood exchange, cyclophosphamide pulse therapy to three of 19 Rh-negative liver, heart, and heart-lung
suppress antibody production, and high-dose gamma transplant recipients who received 3 to 153 units
globulin. The DAT became negative 31 days later. (median, 10) of Rh-positive RBCs at surgery developed
Analysis of IgG heavy-chain allotypes (Gm) in this anti-D at 11–15 days after transplantation. One patient
case produced remarkable findings. Allotypes of the might have had an unusually rapid primary immune
donor and recipient were identical before transplanta- response, and two cases were secondary to previous
tion. Gm typing of the eluate from the patient’s RBCs, exposure by pregnancy. The authors pointed out that
however, revealed numerous allotypes and indicated this is a low rate of rhesus immunization compared
that the antibodies could not have been produced either with what would be expected after transfusion of Rh-
by the liver donor or the patient! The authors concluded positive units to Rh-negative persons who were not
that the antibodies were derived from two or more immunosuppressed. Nevertheless, they advised
persons other than the donor and recipient. Therefore, caution for young females and for patients who might
they suggested that the observed delayed hemolytic have been exposed previously to Rh-positive RBCs by
anemia could have been caused by alloantibodies pro- transfusion or pregnancy. Blomqvist and colleagues374
duced by donor lymphocytes derived from the patient’s reported that after liver transplantation, 9% of patients
multiple transfusions. We suggested a similar hypothe- developed new non-ABO blood group alloantibodies,
sis for the formation of apparent RBC “autoantibodies” presumably of recipient origin.
following transfusion359a (see Chapter 9). This concept is Forsyth and associates375 described a patient who
consistent with observations made by others of patients was group A, Rh negative and who had a negative
with delayed hemolytic transfusion reactions in whom preoperative antibody screening test. Intraoperatively,
a positive DAT has persisted for a longer period of time he was given 31 units of group A, Rh-positive RBCs
than the transfused cells could have survived.360,361 This and 46 units of platelets from group A, Rh-positive
phenomenon has previously generally been explained donors. The liver donor’s blood was group O, Rh pos-
on the basis of autoantibody production. itive. On day +5, anti-D and anti-E were detected, and
If the results of Ishikura and colleagues can be the patient developed marked hemolysis complicated
confirmed, this will support a new concept regarding by acute renal failure. He required transfusion of 14
alloantibody production after blood transfusion. units of group O, Rh-negative RBCs before the hemo-
Consistent with their data are multiple reports indicat- lysis ceased and his blood count stabilized. The
ing survival of donor lymphocytes after blood transfu- authors suggested that this was probably a secondary
sion, although survival has generally been for a short
period of time.362-365 More persistent microchimerism
might occur after transfusion, however, and could be Immune competence of recipient
responsible for significant immunologic effects. Dzik366
has proposed that microchimerism be viewed as
occupying a middle position on a spectrum between GVHD
HLA similarity of donor and recipient

alloimmunization and GVHD (Figure 12-19). Immuno-


logically normal persons can be expected to respond to
exposure to allogeneic leukocytes by developing HLA
antibodies. At the other extreme are transfusions of non-
Microchimerism
irradiated blood containing viable mononuclear cells,
given to recipients with severe immunodeficiency,
which can result in engraftment, rapid expansion of
donor cells, and GVHD. Between these extremes could
be circumstances in which donor cells are neither
quickly eliminated from the host nor allowed
unchecked clonal expansion. Instead, a balanced state of Alloimmunization
microchimerism might ensue. Indeed, Starzl and associ-
ates367,368 have described a ubiquitous low-level donor-
leukocyte microchimerism in recipients as long as
30 years after solid organ transplantation. Dose of allogeneic WBCs
FIGURE 12-19. Mononuclear cell mircochimerism and the spectrum
Alloantibodies of Recipient Origin between graft-versus-hot disease (GVHD) and HLA alloimmunization.
The development of microchimerism after allogeneic transfusion could
Red cell alloantibodies of host origin are produced by depend on the number of viable donor WBCs transfused, the
immunocompetence of the recipient, and the HLA similarity of donor
some patients after solid organ transplantation in spite and recipient. (From Dzik WH: Mononuclear cell microchimerism and
of the immunosuppressive therapy with which such the immunomodulatory effect of transfusion. Transfusion
patients are treated. Rh-negative recipients of Rh-posi- 1994;34:1007.)
506 Immune Hemolytic Anemias

immune response, although the only known previous increased to 4.6 mg/dL (2.0 mg/dL indirect), and the
exposure to blood occurred several years before uncorrected reticulocyte count increased to 4.4%.
transplantation, when the patient was an intravenous There was no hemoglobinuria or hemoglobinemia,
drug user. and hemolysis resolved in about 10 days.
Hyma and coworkers376 reported on a patient who
developed postoperative RBC alloantibodies (anti-Jka,
-c, and –S) and hemolysis 9 days after liver transplan- Alloimmune Thrombocytopenia
tation. These alloantibodies had not been present in the West and colleagues382 reported three unrelated
recipient’s serum before transplantation or in the sera patients who each received an organ (two kidneys and
of the liver or blood donors. The patient had received a liver) from the same donor. In all three patients,
only one transfusion 10 years before transplantation, severe alloimmune thrombocytopenia developed as a
after which time he was noted to have anti-K. During result of antibodies against the HPA-1a (PLA1)
surgery, the patient was transfused with 10 units of K- alloantigen. In these three patients, the thrombocy-
negative RBCs and 12 units of platelets. The liver topenia was refractory to all medical maneuvers
donor’s red cells were positive for Jka, c, and S antigens. except the transfusion of HPA-1a–negative platelets.
Immunosuppression initially consisted of cyclosporine, In one patient, the thrombocytopenia contributed to
azathioprine, and prednisolone. death. In another, the thrombocytopenia was cured by
Alloimmunization is not frequent in immunosup- splenectomy, and in the third patient, the thrombocy-
pressed solid organ transplant recipients, however. topenia resolved after an episode of severe graft rejec-
Casanueva and colleagues377 reported that 17 Rh-neg- tion. The donor had HPA-1a antibodies in her serum
ative liver transplant patients received 5 to 41 units of despite the passage of more than 20 years since her
Rh-positive red cells during surgery. The patients last pregnancy. The authors emphasized that a diag-
were followed for 7 weeks to 70 months after trans- nosis of transplant-mediated alloimmune thrombocy-
plantation for detection of unexpected antibodies. topenia must be considered in patients with severe
Cyclosporin A and prednisone, azathioprine, and thrombocytopenia after organ transplantation.
adjunctive rabbit antilymphocyte globulin or mono-
clonal OKT3 antibody were used to prevent graft
rejection. None of the patients produced anti-D or any Transmission of Idiopathic
other unexpected red cell alloantibody. Thrombocytopenic Purpura
by Liver Transplantation
Autoimmune Hemolytic Anemia Friend and associates383 described a patient who devel-
Rougier and associates378described a 46-year-old oped severe, life-threatening thrombocytopenia after
female with end-stage renal disease and no history of receiving a liver transplant from a donor with long-
autoimmune disease who received an ABO- and standing idiopathic thrombocytopenic purpura who
ABDR-identical renal allograft from a cadaver donor. had had a fatal intracranial hemorrhage. Antibodies
On day 16 after transplantation, while receiving reactive with the glycoprotein (GP) complex IIb–IIIa
immunosuppressive therapy including antithymocyte were detected in serum from the liver donor, and anti-
globulin, azathioprine, prednisone, and cyclosporine, bodies with the same specificity were identified in the
she developed manifestations of AIHA. The hemoglo- recipient after transplantation. The platelet count was
bin dropped from 8.8 g/dL to 6 g/dL, haptoglobin 217,000/μL on the first postoperative day but fell to
was less than 0.06 g/L, bilirubin increased slightly to 10,000/μL the following day and remained at
16 μmol/L, and hemoglobinuria was present. The extremely low levels until the eighth postoperative day.
DAT was positive (IgM and complement), and no The patient was treated with platelet transfusions, cor-
specificity of the antibody could be determined. ticosteroids, plasma exchange, and intravenous gamma
Hemolysis lasted 45 days before hemoglobin slowly globulin; she had already undergone splenectomy. It
increased after cyclosporine had been greatly was not certain whether these treatments were effec-
reduced. The AIHA had resolved completely at 7 tive; however, the antibodies disappeared and her
months after transplantation. The authors speculated thrombocytopenia resolved after a second liver trans-
that the autoantibodies were produced by donor B plant, which was necessary because of deteriorating
lymphocytes in the recipient. Other cases of AIHA liver function. The authors suggest that the most likely
after ABO-compatible renal transplants have also mechanism to account for the clinical findings was the
been reported.379,380 transfer of autoantibody-producing donor lympho-
Beiting and Larimore381 reported on a 33-year-old cytes in sufficient number to cause autoantibody to
male who developed an autoanti-I with a titer of 512 accumulate in the recipient. The heart from the original
in 30% albumin at 4°C and a maximum thermal liver donor was transplanted into a 42-year-old female
amplitude of 37°C. The patient developed a weakly with idiopathic cardiomyopathy. She had an uncompli-
positive DAT with only complement detectable on the cated recovery with no bleeding and no need for post-
RBCs. His hemoglobin fell to 6.5 g/dL, total bilirubin operative platelet transfusions.
Immune Hemolysis Associated with Transplantation 507

Immune Neutropenia after Renal after allogeneic bone marrow transplantation. Blood
1986;67:177–181.
Transplantation 7. Hazlehurst GR, Brenner MK, Wimperis JZ, Knowles SM,
Prentice HG: Haemolysis after T-cell depleted bone marrow
Jondeau and coworkers384 reported a patient who transplantation involving minor ABO incompatibility. Scand J
developed neutropenia 3 weeks after a cadaveric Haematol 1986;37:1–3.
kidney transplant. In spite of numerous attempts at 8. Klumpp TR: Immunohematologic complications of bone
treatment, the neutropenia persisted until day 97 after marrow transplantation. Bone Marrow Transplant 1991;8:
159–170.
transplantation. An anti-NA1 antibody was found in 9. Klumpp TR, Block CC, Caligiuri MA, Rabinowe SN, Soiffer
the serum, and the patient’s granulocytes were posi- RJ, Ritz J: Immune-mediated cytopenia following bone
tive for this antigen. The authors suggested that the marrow transplantation: Case reports and review of the liter-
antibody was an autoantibody, while recognizing that ature. Medicine 1992;71:73–83.
an alternative possibility was that the antibody was an 10. Rowley S, Braine H: Probable hemolysis following minor
ABO incompatible marrow transplantation (IMT) [abstract].
alloantibody produced by donor-derived passenger B Blood 1982;60(Suppl. 1):171.
lymphocytes. 11. Haas RJ, Rieber P, Helmig M, Strobel E, Belohradsky BH,
Heim MU: Acquired immune haemolysis by anti A 1 antibody
following bone marrow transplantation. Blut 1986;53:401–404.
Microchimerism after Solid Organ 12. Gajewski JL, Petz LD, Calhoun L, et al: Hemolysis of transfused
Transplantation group O red blood cells in minor ABO-incompatible unrelated-
donor bone marrow transplants in patients receiving
Starzl and coworkers367 and Fung368 have described a cyclosporine without posttransplant methotrexate. Blood
ubiquitous low-level donor-leukocyte microchimerism 1992;79:3076–3085.
13. Robertson VM, Henslee PJ, Jennings CD, Hill MG, Thompson
in recipients of solid organ allografts as long as 30 years JT, Dickson LG: Early appearance of anti-A isohemagglutinin
after transplantation. They postulate that the interaction after allogeneic, ABO minor incompatible, T cell depleted
of coexisting donor and recipient leukocyte populations bone marrow transplant. Transplant Proc 1987;19:4612–4617.
leads to the down-regulation of immune responsive- 14. Laurencet FM, Samii K, Bressoud A, et al: Massive delayed
hemolysis following peripheral blood stem cell transplanta-
ness to donor alloantigens by recipient cells and also tion with minor ABO incompatibility. Hematol Cell Ther
causes hyporesponsiveness to recipient alloantigens by 1997;39:159–162.
donor immunocompetent leukocytes. Allograft sur- 15. Rowley SD: Hematopoietic stem cell transplantation between
vival requires an umberella of immunosuppression that red cell incompatible donor-recipient pairs. Bone Marrow
protects both the donor cells (including the allograft) Transplant 2001;28:315–321.
16. Tiplady CW, Fitzgerald JM, Jackson GH, Conn JS, Proctor SJ:
from rejection and the recipient cells from the possibil- Massive haemolysis in a group A recipient of a group O
ity of GVHD. In an attempt to prevent rejection of the peripheral blood stem cell allogeneic transplant. Transfus
allograft, various methods, such as donor adjuvant Med 2001;11:455–458.
bone marrow augmentation, have been used to 17. Sokol RJ, Stamps R, Booker DJ, et al: Posttransplant immune-
mediated hemolysis. Transfusion 2002;42:198–204.
augment the natural microchimerism in clinical trans- 18. Bolan CD, Childs RW, Procter JL, Barrett AJ, Leitman SF:
plantation. The incidence of early acute rejection, Massive immune haemolysis after allogeneic peripheral blood
however, was not different from that observed in stem cell transplantation with minor ABO incompatibility. Br
patients who did not receive donor bone marrow, and J Haematol 2001;112:787–795.
the prevention of chronic rejection by this technique has 19. Toren A, Dacosta Y, Manny N, Varadi G, Or R, Nagler A:
Passenger B-lymphocyte-induced severe hemolytic disease
not yet been documented.368 Indeed, other researchers after allogeneic peripheral blood stem cell transplantation.
question whether microchimerism is the cause or Blood 1996;87:843–844.
merely a consequence of long-term allograft survival.385 20. Greeno EW, Perry EH, Ilstrup SJ, Weisdorf DJ: Exchange
transfusion the hard way: Massive hemolysis following trans-
plantation of bone marrow with minor ABO incompatibility.
Transfusion 1996;36:71–74.
R E F E R E N C E S 21. Oziel-Taieb S, Faucher-Barbey C, Chabannon C, et al: Early
and fatal immune haemolysis after so-called “minor” ABO-
1. Petz LD: Immunohematologic problems associated with bone incompatible peripheral blood stem cell allotransplantation.
marrow transplantation. Transfus Med Rev 1987;1:85–100. Bone Marrow Transplant 1997;19:1155–1156.
2. Petz LD: Hemolysis associated with transplantation. 22. Broketa G, Simpson JK, Hammert L, Ball E: Delayed hemoly-
Transfusion 1998;38:224–228. sis after minor ABO mismatched peripheral blood stem cell
3. Petz LD: Immunohematologic problems unique to bone transplantation. Transfusion 1997;37:37S.
marrow transplantation. In Garratty G (ed): Red Cell Antigens 23. Leo A, Mytilineos J, Voso MT, et al: Passenger lymphocyte
and Antibodies. Arlington, VA: American Association of syndrome with severe hemolytic anemia due to an anti-Jk(a)
Blood Banks, 1986:195–229. after allogeneic PBPC transplantation. Transfusion 2000;
4. Sniecinski IJ, Oien L, Petz LD, Blume KG: Immunohematologic 40:632–636.
consequences of major ABO-mismatched bone marrow trans- 24. Borka P, Jakab J, Rajczy K, et al: Temporary donor-derived B-
plantation. Transplantation 1988;45:530–534. lymphocyte microchimerism leading to hemolysis in minor
5. Petz LD: The expanding boundaries of transfusion medicine. ABO-incompatible renal transplantation. Transplant Proc
In Nance SJ (ed): Clinical and Basic Science Aspects of 2001;33:2287–2289.
Immunohematology. Arlington, VA: American Association of 25. ABO, Lewis, and P groups and Ii antigens. In Mollison PL,
Blood Banks, 1991:73–113. Engelfriet CP, Contreras M (eds): Blood Transfusion
6. Hows J, Beddow K, Gordon Smith E, et al: Donor- in Clinical Medicine, 10th ed. Oxford: Blackwell Science,
derived red blood cell antibodies and immune hemolysis 1997:115–150.
508 Immune Hemolytic Anemias

26. Bakacs T, Tusnady G, Vegh Z, Merry AH: Red-cell bound anti- engraftment and graft-versus-host disease. Blood
A is more efficient than anti-B in competition for fluid phase 1995;86:2842–2848.
complement. Immunol Lett 1993;35:213–218. 47. Pan L, Delmonte J Jr, Jalonen CK, Ferrara JL: Pretreatment of
27. Bakacs T, Vegh Z, Merry AH, et al: Interactions in the comple- donor mice with granulocyte colony-stimulating factor polar-
ment-mediated lysis of blood group AB erythrocytes sensi- izes donor T lymphocytes toward type-2 cytokine production
tized simultaneously with anti-A and anti-B monoclonal and reduces severity of experimental graft-versus-host
antibodies. Immunol Lett 1993;35:219–228. disease. Blood 1995;86:4422–4429.
28. Robertson V, Hill M, Bryan J, Dickson L: Anti-JK(b) identified 48. Lapierre V, Oubouzar N, Auperin A, et al: Influence of the
in JK(b) positive recipient following T-cell depleted bone hematopoietic stem cell source on early immunohematologic
marrow transplant [abstract]. Transfusion 1987;27:525. reconstitution after allogeneic transplantation. Blood
29. Ting A, Pun A, Dodds AJ, Atkinson K, Biggs JC: Red cell 2001;97:2580–2586.
alloantibodies produced after bone marrow transplantation. 49. Mielcarek M, Zaucha JM, Sandmaier B, et al: Type of post-
Transfusion 1987;27:145–147. grafting immunosuppression after non-myeloablative blood
30. Young PP, Goodnough LT, Westervelt P, Diersio JF: Immune cell transplantation may influence risk of delayed haemolysis
hemolysis involving non-ABO/RhD alloantibodies following due to minor ABO incompatibility. Br J Haematol
hematopoietic stem cell transplantation. Bone Marrow 2002;116:500–501.
Transplant 2001;27:1305–1310. 50. Vogelsang G: Pharmacology and use of immunosuppressive
31. Franchini M, De Gironcoli M, Gandini G, et al: Transmission agents after bone marrow transplantation. In Forman SJ,
of an anti-RhD alloantibody from donor to recipient after Blume KG, Thomas ED (eds): Bone Marrow Transplantation.
ABO-incompatible BMT. Bone Marrow Transplant Boston: Blackwell Scientific, 1994:114–123.
1998;21:1071–1073. 51. Kazmers IS, Daddona PE, Dalke AP, Kelley WN: Effect of
32. Myser T, Steedman M, Hunt K, Strohm P, Williams M, immunosuppressive agents on human T and B lymphoblasts.
Kennedy M: A bone marrow transplant with an acquired anti- Biochem Pharmacol 1983;32:805–810.
Le(a): A case study. Hum Immunol 1986;17:102–106. 52. Rosenthal GJ, Weigand GW, Germolec DR, Blank JA, Luster
33. Needs ME, McCarthy DM, Barrett J: ABH and Lewis antigen MI: Suppression of B cell function by methotrexate and trime-
and antibody expression after bone marrow transplantation. trexate: Evidence of purine biosynthesis as a major mecha-
Acta Haematol 1987;78:13–16. nism of action. J Immunol 1988;148:410–416.
34. Gale RP, Feig S, Ho W, Falk P, Rippee C, Sparkes R: ABO blood 53. Bar BM, Santos GW, Donnenberg AD: Reconstitution of anti-
group system and bone marrow transplantation. Blood body response after allogeneic bone marrow transplantation:
1977;50:185–194. Effect of lymphocyte depletion by counterflow centrifugal
35. Oriol R, Le Pendu J, Sparkes RS, et al: Insights into the expres- elutriation on the expression of hemagglutinins. Blood
sion of ABH and lewis antgens through human bone marrow 1990;76:1410–1418.
transplantation. Am J Hum Genet 1981;33:551–560. 54. Lasky LC, Warkentin PI, Kersey JH, Ramsay NK, McGlave PB,
36. Lizza C, Myers J, Gindy L: Blood groups. In Petz LD, Swisher McCullough J: Hemotherapy in patients undergoing blood
SN, Kleinman S, Spence RK, Strauss RG (eds): Clinical group incompatible bone marrow transplantation.
Practice of Transfusion Medicine, 3rd ed. New York: Churchill Transfusion 1983;23:277–285.
Livingstone, 1996:71–151. 55. Inwood MJ, Zuliani B: Anti-A hemolytic transfusion with
37. Bensinger WI, Buckner CD, Thomas ED, Clift RA: ABO- packed O cells. Ann Intern Med 1978;89:515–516.
incompatible marrow transplants. Transplantation 1982;33: 56. Moroff G, Friedman A, Robkin Kline L, Gautier G, Luban NL:
427–429. Reduction of the volume of stored platelet concentrates for
38. Noborio K, Muroi K, Izumi T, et al: Massive immune hemoly- use in neonatal patients. Transfusion 1984;24:144–146.
sis after non-myeloablative allogeneic peripheral blood stem 57. Simon TL, Sierra ER: Concentration of platelet units into small
cell transplantation with minor ABO-incompatibility. Leuk volumes. Transfusion 1984;24:173–175.
Lymphoma 2003;44:357–359. 58. Pineda AA, Zylstra VW, Clare DE, Dewanjee MK, Forstrom
39. Worel N, Greinix HT, Keil F, et al: Severe immune hemolysis LA: Viability and functional integrity of washed platelets.
after minor ABO-incompatible allogeneic peripheral blood Transfusion 1989;29:524.
stem cell transplantation occurs more frequently after non- 59. Sniecinski I, O’Donnell MR: Hemolytic Complications of
myeloablative than myeloablative conditioning. Transfusion Hematopoietic Cell Transplantation. In Thomas ED, Blume
2002;42:1293–1301. KG, Forman SJ (eds): Hematopoietic Cell Transplantation.
40. Herzog P, Korinkova P, Stambergova M, Lukasova M: Auto Malden, MA: Blackwell Science, 1999:674–684.
anti-A1 and auto anti-NA1 after bone marrow transplanta- 60. Friedberg RC: Transfusion therapy in the patient undergoing
tion. Folia Haematol Int Mag Klin Morphol Blutforsch hematopoietic stem cell transplantation. Hematol Oncol Clin
1987;114:874–880. North Am 1994;8:1105–1116.
41. Salmon JP, Michaux S, Hermanne JP, et al: Delayed massive 61. Petz LD: Platelet transfusions. In Petz LD, Swisher SN,
immune hemolysis mediated by minor ABO incompatibility Kleinman S, Spence RK, Strauss RG (eds): Clinical Practice of
after allogeneic peripheral blood progenitor cell transplanta- Transfusion Medicine, 3rd ed. New York: Churchill
tion. Transfusion 1999;39:824–827. Livingstone, 1996:359–412.
42. Bornhauser M, Ordemann R, Paaz U, et al: Rapid engraftment 62. Matsue K, Yasue S, Matsuda T, et al: Plasma glycosyl
after allogeneic ABO-incompatible peripheral blood progeni- transferase activity after ABO-incompatible bone marrow
tor cell transplantation complicated by severe hemolysis. transplantation and development of an inhibitor for glycosyl-
Bone Marrow Transplant 1997;19:295–297. transferase activity. Exp Hematol 1989;17:827–831.
43. Delamaire M, le Berre CI, Dauriac C, et al: Fatal hemolysis 63. Yoshida A, Schmidt GM, Blume KG, Beutler E: Plasma blood
following allogeneic peripheral blood stem cells (PBSC) trans- group glycosyltransferase activities after bone marrow trans-
plantation [abstract]. Vox Sanguinis 1998;74(Suppl.):1533. plantation. Blood 1980;55:699–701.
44. Ramsey G: Red cell antibodies arising from solid organ trans- 64. Maeda K, Taniwaki K, Santo T, et al: Anti-A and/or anti-B is
plants. Transfusion 1991;31:76–86. not detectable in some patients who underwent ABO-incom-
45. Magrin GT, Street AM, Williams TJ, Esmore DS: Clinically patible bone marrow transplantation. Transfusion
significant anti-A derived from B lymphocytes after single 1995;35:635–639.
lung transplantation. Transplantation 1993;56:466. 65. Renton PH, Hancock JA: Uptake of A and B antigens by trans-
46. Korbling M, Huh YO, Durett A, et al: Allogeneic blood stem fused group O erythrocytes. Vox Sang 1962;7:33–38.
cell transplantation: Peripheralization and yield of donor- 66. Crookston MC, Tilley CA: A and B Lewis antigens in plasma.
derived primitive hematopoietic progenitor cells (CD34+ Thy- Proceedings of the Fifth International Convocation on
1dim) and lymphoid subsets, and possible predictors of Immunologics. Basel: Karger, 1977:246–256.
Immune Hemolysis Associated with Transplantation 509

67. Arndt PA, Leger RM, Garratty G, Calhoun L, Chow Y, Petz 86. Sahovic EA, Flick J, Graham CD, Stuart RK: Case report:
LD: Use of flow cytometry for detecting uptake of A/B blood Isoimmune inhibition of erythropoiesis following ABO-
group substance on transfused/transplanted O RBCs incompatible bone marrow transplantation. Am J Med Sci
[abstract]. Transfusion 1999;39(Suppl.):43S–44S. 1991;302:369–373.
68. Buckner CD, Clift RA, Sanders JE, et al: ABO-incompatible 87. Karhi KK, Andersson LC, Vuopio P, Gahmberg CG:
marrow transplants. Transplantation 1978;26:233–238. Expression of blood group A antigens in human bone marrow
69. Lee JH, Lee JH, Choi SJ, et al: Changes of isoagglutinin titres cells. Blood 1981;57:147.
after ABO-incompatible allogeneic stem cell transplantation. 88. Peggs KS, Morris EC, Kottaridis PD, et al: Outcome of major
Br J Haematol 2003;120:702–710. ABO-incompatible nonmyeloablative hematopoietic stem cell
70. Warkentin PI, Yomtovian R, Hurd D, Brunning R: Severe transplantation may be influenced by conditioning regimen.
delayed hemolytic transfusion reaction complicating an ABO- Blood 2002;99:4642–4643.
incompatible bone marrow transplantation. Vox Sang 89. Veelken H, Wasch R, Behringer D, Bertz H, Finke J: Pure red
1983;45:40–47. cell aplasia after allogeneic stem cell transplantation with
71. Witherspoon RP, Storb R, Ochs HD, et al: Recovery of anti- reduced conditioning. Bone Marrow Transplant 2000;26:
body production in human allogeneic marrow graft recipi- 911–915.
ents: Influence of time posttransplantation, the presence or 90. Dinsmore RE, Reich LM, Kapoor N, et al: ABH incompatible
absence of chronic graft-versus-host disease, and antithymo- bone marrow transplantation: Removal of erythrocytes by
cyte globulin treatment. Blood 1981;58:360–368. starch sedimentation. Br J Haematol 1983;54:441–449.
72. Ho WG, Champlin RE, Feig SA, Gale RP: Transplantation of 91. Warkentin PI, Hilden JM, Kersey JH, Ramsay NKC,
ABH incompatible bone marrow: Gravity sedimentation of McCullough J: Transplantation of major ABO-incompatible
donor marrow. Br J Haematol 1984;57:155–162. bone marrow depleted of red cells by hydroxyethyl starch.
73. Lopez J, Steegmann JL, Perez G, et al: Erythopoietin in the Vox Sang 1985;48:89–104.
treatment of delayed immune hemolysis of a major ABO- 92. Braine HG, Sensenbrenner LL, Wright SK, Tutschka PJ,
incompatible bone marrow transplant. Am J Hematol Saral R, Santos GW: Bone marrow transplantation with major
1994;45:237. ABO blood group incompatibility using erythrocyte depletion
74. Biggs JC, Concannon AJ, Dodds AJ, Isbister JP, Kesteven P: of marrow prior to infusion. Blood 1982;60:420–425.
Allogeneic bone marrow transplantation across the ABO 93. Ritchey B, Henry S, Forman SJ, et al: A new technique using
barrier. Med J Aust 1979;2:173–175. differential centrifugation of bone marrow for ABO incompat-
75. Gmur J, Burger J, Schaffner A, et al: Pure red cell aplasia of ible transplants. Transplantation 1983;35:638–639.
long duration complicating major ABO-incompatible bone 94. Sniecinski I, Henry S, Ritchey B, Branch DR, Blume KG:
marrow transplantation. Blood 1990;75:290–295. Erythrocyte depletion of ABO-incompatible bone marrow. J
76. Bar BMAM, Van Dijk BA, Schattenberg A, de Man AJ, Kunst Clin Apheresis 1985;2:231–234.
VA, de Witte T: Erythrocyte repopulation after major ABO 95. Jin N, Hill R, Segal G, et al: Preparation of red blood-cell-
incompatible transplantation with lymphocyte-depleted bone depleted marrow for ABO-incompatible marrow transplanta-
marrow. Bone Marrow Transplant 1995;16:793–799. tion by density-gradient separation using the IBM 2991 blood
77. Blacklock HA, Gilmore MJ, Prentice HG, et al: ABO-incom- cell processor. Exp Hematol 1987;15:93–98.
patible bone-marrow transplantation: Removal of red blood 96. Falkenburg JHF, Schaafsma MR, Jansen J, et al: Recovery
cells from donor marrow avoiding recipient antibody deple- of hematopoiesis after blood-group-incompatible bone
tion. Lancet 1982;1061–1064. marrow transplantation with red-blood-cell-depleted grafts.
78. Hows JM, Chipping PM, Palmer S, Gordon Smith EC: Transplantation 1985;39:514–520.
Regeneration of peripheral blood cells following ABO incom- 97. Chan KW, Unrau L, Denegri JF: ABO incompatible marrow
patible allogeneic bone marrow transplantation for severe transplant. Transfusion 1983;23:80.
aplastic anaemia. Br J Haematol 1983;53:145–151. 98. Hershko C, Gale RP, Ho W, Fitchen J: ABH antigens and bone
79. Bolan CD, Leitman SF, Griffith LM, et al: Delayed donor red marrow transplantation. Br J Haematol 1980;44:65–73.
cell chimerism and pure red cell aplasia following major ABO- 99. Reviron J, Schenmetzler C, Bussel A, Frappaz D, Devergie A,
incompatible nonmyeloablative hematopoietic stem cell Gluckman E: Obstacle to red cell engraftment due to major
transplantation. Blood 2001;98:1687–1694. ABO incompatibility in allogeneic bone marrow transplants
80. Yang MH, Hsu HC: Pure red cell aplasia after ABO-incompat- (BMT): Quantitative and kinetic aspects in 58 BMTs.
ible allogeneic stem cell transplantation in severe aplastic Transplant Proc 1987;19:4618–4622.
anemia with response to steroids: A case report and literature 100. Reviron J, Schenmetzler C, Bussel A, Devergie A, Gluckman E:
review. Ann Hematol 2001;80:299–301. Evidence for different kinds of major ABO incompatibility in
81. Santamaria A, Sureda A, Martino R, Domingo-Albos A, transplantation due to the interplay of qualitative and quanti-
Muniz-Diaz E, Brunet S: Successful treatment of pure red cell tative factors: Application to the management of 62 bone
aplasia after major ABO-incompatible T cell-depleted bone marrow recipients. Transplant Proc 1987;19:4623–4628.
marrow transplantation with erythropoietin. Bone Marrow 101. Volin l, Ruutu T: Pure red cell aplasia of long duration after
Transplant 1997;20:1105–1107. major ABO-incompatible bone marrow transplantation. Acta
82. Bierman PJ, Warkentin P, Hutchins MR, Klassen LW: Pure red Haematol 1990;84:195–197.
cell aplasia following ABO mismatched marrow transplanta- 102. Labar B, Bogdanic V, Nemet D, et al: Antilymphocyte globu-
tion for chronic lymphocytic leukemia: Response to antithy- lin for treatment of pure red cell aplasia after major ABO
mocyte globulin. Leuk Lymphoma 1993;9:169–171. incompatible marrow transplant. Bone Marrow Transplant
83. Benjamin RJ, Connors JM, McGurk S, Churchill WH, Antin 1992;10:471–472.
JH: Prolonged erythroid aplasia after major ABO-mismatched 103. Or R, Naparstek E, Mani N, Slavin S: Treatment of pure red-
transplantation for chronic myelogenous leukemia. Biol Blood cell aplasia following major ABO-mismatched T-cell-depleted
Marrow Transplant 1998;4:151–156. bone marrow transplantation: Two case reports with success-
84. Marmont AM, Damasio EE, Bacigalupo A, et al: A to O bone ful response to plasmapheresis. Transplant Int 1991;4:99.
marrow transplantation in severe aplastic anaemia: Dynamics 104. Ohashi K, Akiyama H, Takamoto S, Tanikawa S, Sakamaki H,
of blood group conversion and demonstration of early Onozawa Y: Treatment of pure red cell aplasia after major
dyserythropoiesis in the engrafted marrow. Br J Haematol ABO-incompatible bone marrow transplantation resistant to
1977;36:511–518. erythropoietin. Bone Marrow Transplant 1994;13:335–336.
85. Blacklock HA, Katz F, Michalevicz R, et al: A and B blood 105. Heyll A, Aul C, Runde V, Arning M, Schneider W, Wernet P:
group antigen expression on mixed colony cells and erythroid Treatment of pure red cell aplasia after major ABO-incompati-
precursors: Relevance for human allogeneic bone marrow ble bone marrow transplantation with recombinant erythropoi-
transplantation. Br J Haematol 1984;58:267–276. etin. Blood 1991;77:906.
510 Immune Hemolytic Anemias

106. Paltiel O, Cournoyer D, Rybka W: Pure red cell aplasia fol- and cyclophosphamide. Bone Marrow Transplant 1995;15:
lowing ABO-incompatible bone marrow transplantation: 105–110.
Response to erythropoietin. Transfusion 1993;33:418–421. 125. Bensinger W, Petersen FB, Banaji M, et al: Engraftment and
107. Sniecinski I: Management of ABO incompatibility in allo- transfusion requirements after allogeneic marrow trans-
geneic bone marrow transplantation. In Forman SJ, Blume plantation for patients with acute non-lymphocytic leukemia
KG, Thomas ED (eds): Bone Marrow Transplantation. Oxford: in first complete remission. Bone Marrow Transplant 1989;
Blackwell Scientific, 1994:497–503. 4:409–414.
108. Selleri C, Raiola A, De Rosa G, et al: CD34+-enriched donor 126. Mielcarek M, Torok-Storb B, Storb R: ABO incompatibility and
lymphocyte infusions in a case of pure red cell aplasia and late relapse risk in patients undergoing allogeneic marrow trans-
graft failure after major ABO-incompatible bone marrow plantation for acute myeloid leukemia. Bone Marrow
transplantation. Bone Marrow Transplant 1998;22:605–607. Transplant 2002;30:547–548.
109. Bavaro P, Di Girolamo G, Olioso P, et al: Donor lymphocyte 127. Badros A, Tricot G, Toor A, et al: ABO mismatch may affect
infusion as therapy for pure red cell aplasia following bone engraftment in multiple myeloma patients receiving nonmye-
marrow transplantation. Br J Haematol 1999;104:930–931. loablative conditioning. Transfusion 2002;42:205–209.
110. Yamaguchi M, Sakai K, Murata R, Ueda M: Treatment of 128. Klumpp TR, Caligiuri MA, Rabinowe SN, Soiffer RJ, Murray
pure red cell aplasia after major ABO-incompatible peri- C, Ritz J: Autoimmune pancytopenia following allogeneic
pheral blood stem cell transplantation by induction of bone marrow transplantation. Bone Marrow Transplant
chronic graft-versus-host disease. Bone Marrow Transplant 1990;6:445–447.
2002;30:539–541. 129. Bashey A, Owen I, Lucas GF, et al: Late onset immune pancy-
111. Mielcarek M, Leisenring W, Torok-Storb B, Storb R: Graft- topenia following bone marrow transplantation. Br J Haemat
versus-host disease and donor-directed hemagglutinin titers 1991;78:268–274.
after ABO-mismatched related and unrelated marrow allo- 130. Sokal E, Michel M, Ninane J, Latinne D, de Bruyere M, Cornu
grafts: Evidence for a graft-versus-plasma cell effect. Blood G: Bone marrow transplantation from an unrelated donor for
2000;96:1150–1156. Fanconi’s anaemia: Two unusual complications. Bone Marrow
112. Friedberg RC: Transfusion therapy in the patient undergoing Transplant 1987;2:99–102.
hematopoietic stem cell transplantation. Hematol Oncol Clin 131. Drobyski WR, Potluri J, Sauer D, Gottschall JL: Autoimmune
N Am 1994;8:1105–1116. hemolytic anemia following T cell-depleted allogeneic bone
113. The Trial to Reduce Alloimmunization and Refractoriness to marrow transplantation. Bone Marrow Transplant 1996;
Platelets Study Group. Leukocyte reduction and ultraviolet B 17:1093–1099.
irradiation of platelets to prevent alloimmunization and 132. Chen FE, Owen I, Savage D, et al: Late onset haemolysis and
refractoriness to platelet transfusions. N Engl J Med 1997; red cell autoimmunisation after allogeneic bone marrow
337:1861–1869. transplant. Bone Marrow Transplant 1997;19:491–495.
114. Sullivan KM: Graft-versus-host disease. In Thomas ED, Blume 133. Wennerberg A, Backman KA, Gillerlain C, Robertson V,
KG, Forman SJ (eds): Hematopietic Cell Transplantation. Joyner T: Mixed erythrocyte chimerism: Implications for
Malden, MA: Blackwell Science, 1999:515–536. tolerance of the donor immune system to recipient non-
115. Storb R, Thomas ED, Weiden PL, et al: Aplastic anemia treated ABO system red cell antigens. Bone Marrow Transplant
by allogeneic bone marrow transplantation: A report on 49 1996;18:433–435.
new cases from Seattle. Blood 1976;48:817–841. 134. Pratt G, Kinsey SE: Remission of severe, intractable auto-
116. Curtis JE, Messner HA: Bone marrow transplantation for immune haemolytic anaemia following matched unrelated
leukemia and aplastic anemia: Management of ABO incom- donor transplantation. Bone Marrow Transplant 2001;
patibility. CMA J 1982;126:649. 28:791–793.
117. Bacigalupo A, Van Lint MT, Occhini D, et al: ABO compatibility 135. Graze PR, Gale RP: Chronic graft versus host disease: a syn-
and acute graft-versus-host disease following allogeneic bone drome of disordered immunity. Am J Med 1979;66:611–620.
marrow transplantation. Transplantation 1988;45:1091–1094. 136. Godder K, Pati AR, Abhyankar SH, Lamb LS, Armstrong W,
118. Barone B, Cordeiro de Paiva TM, da Silva Bouzas LF, et al: Henslee-Downey PJ: De novo chronic graft-versus-host
ABO system incompatibility and graft versus host disease disease presenting as hemolytic anemia following partially
(GVHD) frequency in bone marrow transplanted patients mismatched related donor bone marrow transplant. Bone
[abstract]. Blood 2001;98:374b. Marrow Transplant 1997;19:813–817.
119. Stussi G, Seebach L, Muntwyler J, Schanz U, Gmur J, Seebach 137. Tamura T, Kanamori H, Yamazaki E, et al: Cold agglutinin
JD: Graft-versus-host disease and survival after ABO-incom- disease following allogeneic bone marrow transplantation.
patible allogeneic bone marrow transplantation: A single- Bone Marrow Transplant 1994;13:321–323.
centre experience. Br J Haematol 2001;113:251–253. 138. Au WY, Lie AK, Liang R, Lam CC, Kwong YL: Fulminant late
120. Stussi G, Muntwyler J, Passweg JR, et al: Consequences of onset cold hemagglutinin disease after allogeneic bone marrow
ABO incompatibility in allogeneic hematopoietic stem cell transplantation [e-letters]. Haematologica 2000;85:E10.
transplantation. Bone Marrow Transplant 2002;30:87–93. 139. Azuma E, Nishihara H, Hanada M, et al: Recurrent cold
121. Benjamin RJ, McGurk S, Ralston MS, Churchill WH, Antin JH: hemagglutinin disease following allogeneic bone marrow
ABO incompatibility as an adverse risk factor for survival transplantation successfully treated with plasmapheresis, cor-
after allogeneic bone marrow transplantation. Transfusion ticosteroid and cyclophosphamide. Bone Marrow Transplant
1999;39:179–187. 1996;18:243–246.
121a. Worel N, Kalhs P, Keil F, et al: ABO mismatch increases trans- 140. De Lord C, Marsh JC, Smith JG, Singer CR, Gordon-Smith EC:
plant-related morbidity and mortality in patients given non- Fatal autoimmune pancytopenia following bone marrow
myeloablative allogeneic HPC transplantation. Transfusion transplantation for aplastic anaemia. Bone Marrow Transplant
2003;43:1153–1161. 1996;18:237–239.
122. Daly AS, Xenocostas A, Lipton JH: Transplantation-associated 141. Keung YK, Cobos E, Bolanos-Meade J, Issarachai S, Brideau A,
thrombotic microangiopathy: Twenty-two years later. Bone Morgan D: Evans syndrome after autologous bone marrow
Marrow Transplant 2002;30:709–715. transplant for recurrent Hodgkin’s disease. Bone Marrow
123. Geller RB, Saral R, Piantadosi S, et al: Allogeneic bone marrow Transplant 1997;20:1099–1101.
transplantation after high-dose busulfan and cyclophos- 142. Hartert A, Willenbacher W, Gunzelmann S, et al: Successful
phamide in patients with acute nonlymphocytic leukemia. treatment of thrombocytopenia and hemolytic anemia with
Blood 1989;73:2209–2218. IvIG in a patient with lupus-like syndrome after mismatched
124. Kalaycioglu M, Copelan E, Avalos B, Klein J, Goormastic M, related PBSCT. Bone Marrow Transplant 2001;27:337–340.
Bolwell B: Survival after ABO-incompatible allogeneic bone 143. Sevilla J, Gonzalez-Vicent M, Madero L, Diaz MA: Acute
marrow transplant after a preparative regimen of busulfan autoimmune hemolytic anemia following unrelated cord blood
Immune Hemolysis Associated with Transplantation 511

transplantation as an early manifestation of chronic graft- 163. Abou-Elella AA, Camarillo TA, Allen MB, et al: Low incidence
versus-host disease. Bone Marrow Transplant 2001;28:89–92. of red cell and HLA antibody formation by bone marrow
144. Horn B, Viele M, Mentzer W, Mogck N, DeSantes K, Cowan transplant patients. Transfusion 1995;35:931–935.
M: Autoimmune hemolytic anemia in patients with SCID 164. Chan EYT, Lawton JWM, Lie AKW, Lau CS: Autoantibody
after T cell-depleted BM and PBSC transplantation. Bone formation after allogeneic bone marrow transplantation:
Marrow Transplant 1999;24:1009–1013. Correlation with the reconstitution of CD5+ B cells and
145. Dovat S, Roberts RL, Wakim M, Stiehm ER, Feig SA: Immune occurrence of graft-versus-host disease. Pathology 1997;
thrombocytopenia after umbilical cord progenitor cell trans- 29:184–188.
plant: Response to vincristine. Bone Marrow Transplant 165. Gluckman E, Anderson E, Dausset J: Autolymphocytotoxic
1999;24:321–323. antibodies after bone-marrow transplantation [letter]. Lancet
146. Mccann SR, Lawler M: Mixed chimaerism; detection and 1977;2:146–147.
significance following BMT: Bone Marrow Transplant 166. Gluckman JC, Gluckman E, Azogui O, et al: Monocytotoxic
1993;11:91–94. antibodies after bone marrow transplantation in aplastic
147. Branch DR, Gallagher MT, Forman SJ, Winkler KJ, Petz LD, anemia. Transplantation 1982;33:599–602.
Blume KG: Endogenous stem cell repopulation resulting in 167. Rouquette-Gally AM, Boyeldieu D, Prost AC, Gluckman E:
mixed hematopoietic chimerism following total body irradia- Autoimmunity after allogeneic bone marrow transplantation:
tion and marrow transplantation for acute leukemia. A study of 53 long-term-surviving patients. Transplantation
Transplantation 1982;34:226. 1988;46:238–240.
148. Petz LD: Endogenous stem cell repopulation after high-dose 168. Minchinton RM, Waters AH, Kendra J, Barrett AJ: Auto-
pretransplant radiochemotherapy. Transplant Proc 1985;17:432. immune thrombocytopenia acquired from an allogeneic bone-
149. Petz LD: The use of the term chimaera in bone marrow trans- marrow graft. Lancet 1982;2:627–629.
plantation. Bone Marrow Transplant 1989;4:139–140. 169. Minchinton RM, Waters AH: Autoimmune thrombocytopenia
150. Petz LD: Documentation of engraftment and characteriza- and neutropenia after bone marrow transplantation [letter].
tion of chimerism following marrow transplantation. In Blood 1985;66:752.
Forman SJ, Blume KG, Donnall Thomas E (eds): Bone 170. Waters AH, Metcalfe P, Minchinton RM, Barrett AJ, James DC:
Marrow Transplantation. Boston: Blackwell Scientific, Autoimmune thrombocytopenia acquired from allogeneic
1994:136–148. bone-marrow graft: Compensated thrombocytopenia in bone
151. Petz LD, Yam P, Wallace RB, et al: Mixed hematopoietic marrow donor and recipient [letter]. Lancet 1983;2:1430.
chimerism following bone marrow transplantation for hema- 171. Minchinton RM, Waters AH, Malpas JS, Starke I, Kendra JR,
tologic malignancies. Blood 1987;70:1331–1337. Barrett AJ: Platelet- and granulocyte-specific antibodies after
152. Przepiorka D, Thomas ED, Durnam DM, Fisher L: Use of a allogeneic and autologous bone marrow grafts. Vox Sang
probe to repeat sequence of the Y chromosome for detection of 1984;46:125–135.
host cells in peripheral blood of bone marrow transplant 172. Bierling P, Cordonnier C, Fromont P, et al: Acquired autoim-
recipients. Hematopathology 1991;95:201–206. mune thrombocytopenia after allogeneic bone marrow trans-
153. Negrin RS, Cleary ML: Laboratory evaluation of minimal plantation. Brit J Haemat 1985;59:643–646.
residual disease. In Forman SJ, Blume KG, Donnall Thomas E 173. Benda H, Panzer S, Kiefel V, et al: Identification of the target
(eds): Bone Marrow Transplantation. Boston: Blackwell platelet glycoprotein in autoimmune thrombocytopenia
Scientific, 1994:179–188. occurring after allogeneic bone marrow transplantation. Blut
154. Esteve J, Alcorta I, Pereira A, Carreras E, Rozman C: Anti-D 1989;58:151–153.
antibody of exclusive IgM class after minor Rh(D)- 174. Rappeport J, Mihm M, Reinherz E, Copanski S, Parkman R:
mismatched BMT. Bone Marrow Transplant 1995;16:632–633. Acute graft versus host disease in recipients of bone marrow
155. Heim MU, Schleuning M, Eckstein R, et al: Rh antibodies transplants from identical twin donors. Lancet 1979;ii:717–720.
against the pretransplant red cells following Rh- incompatible 175. Spruce W, Forman S, McMillan R, Farbstein M, Turner M,
bone marrow transplantation. Transfusion 1988;28:272–275. Blume K: Idiopathic thrombocytopenic purpura following
156. Niethammer D, Bienzle U, Rodt H, et al: Rhesus incompati- bone marrow transplantation. Acta Haematol 1983;69:47–51.
bility and aplastic anemia as the consequence of split 176. Plouvier E, Herve P, Faivre L, Noir A: Refractory autoimmune
chimerism after bone-marrow transplantation for severe com- thrombocytopenia following autologous stem cell grafting.
bined immunodeficiency. Thymus 1980;2:75–82. Exp Hematol 1983;11:172.
157. Girelli G, Arcese W, Bianchi A, et al: Hemolysis in Rh-negative 177. First LR, Smith BR, Lipton J, Nathan DG, Parkman R,
female recipient after Rh-incompatible bone marrow trans- Rappeport JM: Isolated thrombocytopenia after allogeneic
plantation for chronic myeloid leukemia. Haematologica bone marrow transplantation: Existence of transient and
1986;71:46–49. chronic thrombocytopenic syndromes. Blood 1985;65:368–374.
158. Moore SB, Motschman TL, Dewald GW, Letendre L, Smithson 178. Anasetti C, Rybka W, Sullivan KM, Banaji M, Slichter SJ:
WA, Hoagland HC: A case of unusually early but temporary Graft-v-host disease is associated with autoimmune-like
humoral immune reconstitution after bone marrow transplan- thrombocytopenia. Blood 1989;73:1054–1058.
tation. Transplantation 1985;39:675–677. 179. Bierling P, Pignon JM, Kuentz M, et al: Thrombocytopenia
159. Izumi N, Fuse I, Furukawa T, et al: Long-term production of after bone marrow transplantation caused by a recipient
pre-existing alloantibodies to E and c after allogenic BMT in a origin Br(a) allo-antibody: Presence of mixed chimerism 3
patient with aplastic anemia resulting in delayed hemolytic years after the graft without hematologic relapse. Blood
anemia. Transfusion 2003;43:241–245. 1994;83:274–279.
160. von Tol MJD, Gerritsen EJA, de Lange GG, et al: The origin of 180. Panzer S, Kiefel V, Bartram CR, et al: Immune thrombocy-
IgG production and homogeneous IgG components after allo- topenia more than a year after allogeneic marrow transplan-
geneic bone marrow transplantation. Blood 1996;87:818–826. tation due to antibodies against donor platelets with anti-PlA1
161. Korver K, de Lange GG, van den Bergh RL, et al: Lymphoid specificity: Evidence for a host-derived immune reaction. Brit
chimerism after allogeneic bone marrow transplantation: Y- J Haematol 1989;71:259–264.
chromatin staining of peripheral T and B lymphocytes and 181. Szymanski L, Sahlstedt L, Ruutu T, Jarventie G, Kekomaki R:
allotyping of serum immunoglobulins. Transplantation Persistence of an antibody to the human platelet antigen HPA-
1987;44:643–650. 5b (Br(a)) after successful bone marrow transplantation (BMT)
162. Yam PY, Petz LD, Knowlton RG, et al: Use of DNA restriction [abstract]. Blood 1992;80(Suppl. 1):217a.
fragment length polymorphisms to document marrow 182. Minchinton RM, Waters AH, Malpas JS, Gordon Smith EC,
engraftment and mixed hematopoietic chimerism following Barrett AJ: Selective thrombocytopenia and neutropenia
bone marrow transplantation. Transplantation 1987;43: occurring after bone marrow transplantation—evidence of an
399–407. auto-immune basis. Clin Lab Haematol 1984;6:157–163.
512 Immune Hemolytic Anemias

183. Klumpp TR, Herman JH, Macdonald JS, Schnell MK, 206. Salama A, Mueller Eckhardt C, Kiefel V: Effect of intravenous
Mullaney M, Mangan KF: Autoimmune neutropenia follow- immunoglobulin in immune thrombocytopenia. Lancet
ing peripheral blood stem cell transplantation. Am J Hematol 1983;2:193–195.
1992;41:215–217. 207. Robertson VM, Dickson LG, Romond EH, Ash RC: Positive
184. Koeppler H, Goldman JM: “Auto”-immune neutropenia antiglobulin tests due to intravenous immunoglobulin in
after allogeneic bone marrow transplantation unresponsive patients who received bone marrow transplant. Transfusion
to conventional immunosuppression but resolving promptly 1987;27:28–31.
after splenectomy. Eur J Haematol 1988;41:182–185. 208. Copelan EA, Strohm PL, Kennedy MS, Tutschka PJ:
185. Stroncek DF, Shapiro RS, Filipovich AH, Plachta LB, Clay ME: Hemolysis following intravenous immune globulin therapy.
Prolonged neutropenia resulting from antibodies to neu- Transfusion 1986;26:410–412.
trophil- specific antigen NB1 following marrow transplanta- 209. Thomas MJ, Misbah SA, Chapel HM, Jones M, Elrington G,
tion. Transfusion 1993;33:158–163. Newsom Davis J: Hemolysis after high-dose intravenous Ig
186. Warkentin PI, Clay ME, Kersey JH, Ramsay NK, McCullough [letter]. Blood 1993;82:3789.
J: Successful engraftment of NA1 positive bone marrow in a 210. Nicholls MD, Cummins JC, Davies VJ, Greenwood JK:
patient with a neutrophil antibody, anti-NA1. Hum Immunol Haemolysis induced by intravenously-administered
1981;2:173–184. immunoglobulin. Med J Aust 1989;150:404–406.
187. Tosi P, Bandini G, Tazzari P, Raspadori D, et al: Autoimmune 211. Nakamura S, Yoshida T, Ohtake S, Matsuda T: Hemolysis due
neutropenia after unrelated bone marrow transplantation. to high-dose intravenous gammaglobulin treatment for
Bone Marrow Transplant 1994;14:1003–1004. patients with idiopathic thrombocytopenic purpura. Acta
188. Kim HC, Park CL, Cowan JH, III, Fattori FD, August CS: Haematol 1986;76:115–118.
Massive intravascular hemolysis associated with intravenous 212. Okubo S, Ishida T, Yasunaga K: Hemolysis after intravenous
immunoglobulin in bone marrow transplant recipients. Am J immune globulin therapy: Relation to IgG subclasses of red
Pediatr Hematol Oncol 1988;10:69–74. cell antibody. Transfusion 1990;30:436–438.
189. Murphy MF, Hook S, Waters AH, et al: Acute haemolysis 213. Brox AG, Cournoyer D, Sternbach M, Spurll G: Hemolytic
after ABO-incompatible platelet transfusions. Lancet anemia following intravenous gamma globulin administra-
1990;335:974–975. tion. Am J Med 1987;82:633–635.
190. Zoes C, Dube VE, Miller HJ, Vye MV: Anti-A1 in the plasma 214. Nakagawa M, Watanabe N, Okuno M, Kondo M, Okagawa H,
of platelet concentrates causing a hemolytic reaction. Taga T: Severe hemolytic anemia following high-dose intra-
Transfusion 1977;17:29–32. venous immunoglobulin administration in a patient with
191. McLeod BC, Sassetti RJ, Weens JH, Vaithianathan T: Haemolytic Kawasaki disease. Am J Hematol 2000;63:160–161.
transfusion reaction due to ABO incompatible plasma in a 215. Moscow JA, Casper AJ, Kodis C, Fricke WA: Positive direct
platelet concentrate. Scand J Haematol 1982;28:193–196. antiglobulin test results after intravenous immune globulin
192. Conway LT, Scott EP: Acute hemolytic transfusion reaction administration. Transfusion 1987;27:248–249.
due to ABO incompatible plasma in a plateletapheresis con- 216. Niosi P, Lundberg J, McCullough J, Park BH, Biggar DW:
centrate. Transfusion 1984;24:413–414. Blood group antibodies in human immune serum globulin. N
193. Pierce RN, Reich LM, Mayer K: Hemolysis following platelet Engl J Med 1971;285:1435–1436.
transfusions from ABO-incompatible donors. Transfusion 217. Inwood MJ, Zuliani BH: Hemolytic transfusion reaction [cor-
1985;25:60–62. respondence]. Ann Intern Med 1979;90:278.
194. Ferguson DJ: Acute intravascular hemolysis after a platelet 218. Silvergleid AJ: Hemolytic transfusion reaction [letter]. Ann
transfusion. Can Med Assoc J 1988;138:523–524. Intern Med 1979;90:278.
195. Reis MD, Coovadia AS: Transfusion of ABO-incompatible 219. Salama A, Kiefel V, Amberg R, Mueller Eckhardt C: Treatment
platelets causing severe haemolytic reaction. Clin Lab Hae- of autoimmune thrombocytopenic purpura with rhesus anti-
matol 1989;11:237–240. bodies [anti-Rh0(D)]. Blut 1984;49:29–35.
196. McManigal S, Sims KL: Intravascular hemolysis secondary to 220. Barbolla L, Nieto S, Llamas P, et al: Severe immune haemolytic
ABO incompatible platelet products. An underrecognized anemia caused by intravenous immunoglobulin anti-D in the
transfusion reaction. Am J Clin Pathol 1999;111:202–206. treatment of autoimmune thrombocytopenia. Vox Sang
197. Chow MP, Yung CH, Hu HY, Tzeng CH: Hemolysis after 1993;64:184–185.
ABO-incompatible platelet transfusions. Chung Hua I Hsueh 221. Salama A, Kiefel V, Mueller Eckhardt C: Effect of IgG anti-
Tsa Chih (Taipei) 1991;48:131–134. Rho(D) in adult patients with chronic autoimmune thrombo-
198. Lundberg WB, McGinniss MH: Hemolytic transfusion reac- cytopenia. Am J Hematol 1986;22:241–250.
tion due to anti-A. Transfusion 1975;15:1–9. 222. Gaines AR: Acute onset hemoglobinemia and/or hemoglo-
199. Larsson LG, Welsh VJ, Ladd DJ: Acute intravascular hemoly- binuria and sequelae following Rh(o)(D) immune globulin
sis secondary to out-of-group platelet transfusion. Trans- intravenous administration in immune thrombocytopenic
fusion 2000;40:902–906. purpura patients. Blood 2000;95:2523–2529.
200. Mair B, Benson K: Evaluation of changes in hemoglobin levels 223. Brain MC, Dacie JV, Hourihane DO: Microangiopathic
associated with ABO-incompatible plasma in apheresis haemolytic anemia: The possible role of vascular lesions in
platelets. Transfusion 1998;38:51–55. pathogenesis. Br J Haematol 1962;8:358.
201. Ferguson DJ: Acute intravascular hemolysis after a platelet 224. Brain MC, Hourihane DO: Microangiopathic haemolytic
transfusion. CMAJ 1988;138:523–524. anaemia: The occurrence of haemolysis in experimentally pro-
202. Shanwell A, Ringden O, Wiechel B, Rumin S, Akerblom O: A duced vascular disease. Br J Haematol 1967;13:135–142.
study of the effect of ABO incompatible plasma in platelet 225. Brain MC, Esterly JR, Beck EA: Intravascular haemolysis with
concentrates transfused to bone marrow transplant recipients. experimentally produced vascular thrombi. Br J Haematol
Vox Sang 1991;60:23–27. 1967;13:868–891.
203. Rowe JM, Ciobanu N, Ascensao J, et al: Recommended 226. Bull BS, Rubenberg ML, Dacie JV, Brain MC: Microangio-
guidelines for the management of autologous and allogeneic pathic haemolytic anaemia: Mechanisms of red-cell fragmen-
bone marrow transplantation: A report from the Eastern tation: In vitro studies. Br J Haematol 1968;14:643–652.
Cooperative Oncology Group (ECOG). Ann Intern Med 227. Brain MC: Microangiopathic hemolytic anemia [review]. Ann
1994;120:143–158. Rev Med 1970;21:133–143.
204. Oberman HA, Beck ML: Red blood cell sensitization due to 228. Rosner F, Rubenberg ML: Erythrocyte fragmentation in con-
unexpected Rh antibodies in immune serum globulin. sumption coagulopathy. N Engl J Med 1969;280:219–220.
Transfusion 1971;11:382–384. 229. Scully RE, Mark EJ, McNeely WF, McNeely BU: Case records
205. Lang GE, Veldhuis B: Immune serum globulin—a cause for anti- of the Massachusetts General Hospital. N Engl J Med
Rh (D) passive sensitization. Am J Clin Pathol 1973;60:205–207. 1994;331:661–667.
Immune Hemolysis Associated with Transplantation 513

230. Brain MC: Microangiopathic hemolytic anemia [review]. 253. Holler E, Kolb HJ, Hiller E, et al: Microangiopathy in patients
N Engl J Med 1969;281:833–835. on cyclosporine prophylaxis who developed acute graft-
231. Ludmerer KM, Kissane JM: Microangiopathic anemia and dis- versus-host disease after HLA-identical bone marrow trans-
seminated intravascular coagulation in an elderly woman plantation. Blood 1989;73:2018–2024.
(clinicopathologic conference). Am J Med 1983;74:1052–1060. 254. Rabinowe SN, Soiffer RJ, Tarbell NJ, et al: Hemolytic-uremic
232. Alpert LI, Benisch B: Hemangioendothelioma of the liver syndrome following bone marrow transplantation in adults
associated with microangiopathic hemolytic anemia: Report for hematologic malignancies. Blood 1991;77:1837–1844.
of four cases. Am J Med 1970;48:624–628. 255. Pettitt AR, Clark RE: Transplantation-associated thrombotic
233. Rodriguez Erdmann F, Button L, Murray JE, Moloney WC: thrombocytopenic purura and hemolytic uremic syndrome.
Kasabach-Merritt syndrome: Coagulo-analytical observa- Semin Hematol 1997;34:126–133.
tions. Am J Med Sci 1971;261:9–15. 256. Fehr J, Knob M: Comparison of red cell creatine level and
234. Westphal RG, Azen EA: Microangiopathic hemolytic anemia reticulocyte count in appraising the severity of hemolytic
due to congenital cardiovascular anomalies. JAMA processes. Blood 1979;53:966–976.
1971;216:1477–1478. 257. Schriber JR, Herzig GP: Transplantation-associated throm-
235. Solanki DL, Sheikh MU: Fragmentation hemolysis in idio- botic thrombocytopenic purpura and hemolytic uremic syn-
pathic hypertrophic subaortic stenosis. Southern Med J drome. Semin Hematol 1997;34:126–133.
1978;5:599–601. 258. Ohno E, Ohtsuka E, Iwashita T, et al: Hemolytic uremic syn-
236. Westring DW: Aortic valve disease and hemolytic anemia. drome following autologous peripheral blood stem cell trans-
Ann Intern Med 1966;65:203–215. plantation in a patient with malignant lymphoma. Bone
237. Myhre E, Rasmussen K: Mechanical hemolysis in aortic valvu- Marrow Transplant 1997;19:1045–1047.
lar disease and aortic ball-valve prosthesis. Acta Med Scand 259. van der Lelie H, Baars JW, Rodenhuis S, et al: Hemolytic
1969;186:543–547. uremic syndrome after high dose chemotherapy with autolo-
238. Petz LD, Goodman JR: Ringed sideroblasts and intramito- gous stem cell support. Cancer 1995;76:2338–2342.
chondrial iron in cases of mechanical hemolytic anemia: 260. Zeigler ZR, Shadduck RK, Nemunaitis J, Andrews DF,
Significance of these findings and observations on the etiology Rosenfeld CS: Bone marrow transplant-associated thrombotic
of hemolysis. Ann Intern Med 1966;64:635–643. microangiopathy: A case series. Bone Marrow Transplant
239. Petz LD, Catlett JP, Lessin LS: Hemolytic Anemias. In Kelly 1995;15:247–253.
WN (ed): Textbook of Internal Medicine. New York: 261. Gonzalez-Vicent M, Diaz MA, Madero L: An uncommon
Lippincott-Raven, 1997:1454–1472. case of late thrombotic thrombocytopenic purpura (42
240. Elliott MA, Nichols WL: Thrombotic thrombocytopenic months) after autologous peripheral blood stem cell (PBSC)
purpura and hemolytic uremic syndrome. Mayo Clin Proc transplantation in a child. Bone Marrow Transplant
2001;76:1154–1162. 1999;23:735–736.
241. Elliott MA, Nichols WL Jr, Plumhoff EA, et al: 262. Togitani K, Takeyama K, Yokozawa T, et al: Thrombotic
Posttransplantation thrombotic thrombocytopenic purpura: A microangiopathy of hyperacute onset after autologous
single-center experience and a contemporary review. Mayo peripheral blood stem cell transplantation in malignant lym-
Clin Proc 2003;78:421–430. phoma. Bone Marrow Transplant 1998;21:1263–1266.
242. Allford SL, Bird JM, Marks DI: Thrombotic thrombocytopenic 263. Paquette RL, Tran L, Landaw EM: Thrombotic microangiopa-
purpura following stem cell transplantation. Leuk Lymphoma thy following allogeneic bone marrow transplantation is asso-
2002;43:1921–1926. ciated with intensive graft-versus-host disease prophylaxis.
243. George JN, El-Harake M: Thrombocytopenia due to enhanced Bone Marrow Transplant 1998;22:351–357.
platelet destructon by nonimmunologic mechanisms. In 264. Lugassy G, Ducach A, Blumenthal R: Fatal chronic relapsing
Beutler E, Lichtman MA, Coller BS, Kipps TJ (eds): Williams thrombotic thrombocytopenic purpura following autologous
Hematology. New York: McGraw-Hill Health Professions, bone marrow transplantation for chronic myeloid leukemia.
1995:1290–1315. Bone Marrow Transplant 1998;21:859–860.
244. Kelton JG: Thrombotic thrombocytopenic purpura and 265. Kondo M, Kojima S, Horibe K, Kato K, Matsuyama T:
hemolytic uremic syndrome: Will recent insight into pathogen- Hemolytic uremic syndrome after allogeneic or autologous
esis translate into better treatment? Transfusion 2002;42:388–392. hematopoietic stem cell transplantation for childhood malig-
245. George JN: How I treat patients with thrombotic thrombocy- nancies. Bone Marrow Transplant 1998;21:281–286.
topenic purpura-hemolytic uremic syndrome. Blood 2000;96: 266. Sarkodee-Adoo C, Sotirescu D, Sensenbrenner L, et al:
1223–1229. Thrombotic microangiopathy in blood and marrow transplant
246. Rock G, Shumak K, Kelton J, et al: Thrombotic thrombocy- patients receiving tacrolimus or cyclosporine A. Transfusion
topenic purpura: Outcome in 24 patients with renal impair- 2003;43:78–84.
ment treated with plasma exchange. Canadian Apheresis 267. Chappell ME, Keeling DM, Prentice HG, Sweny P: Haem-
Study Group. Transfusion 1992;32:710–714. olytic uraemic syndrome after bone marrow transplantation:
247. Amorosi EL, Ultmann JE: Thrombotic thrombocytopenic An adverse effect of total body irradiation? Bone Marrow
purpura: Report of 16 cases and review of the literature. Transplantation 1988;3:339–347.
Medicine 1966;45:139. 268. Steele BT, Lirenman PS: Acute radiation nephritis and the
248. Crain SM, Choudhury AM: Thrombotic thrombocytopenic haemolytic uraemic syndrome. Clin Nephrol 1979;11:272.
purpura: A reappraisal. JAMA 1981;246:1243–1246. 269. Krochak RJ, Baker DJ: Radiation nephritis. Clinical manifesta-
249. Zomas A, Saso R, Powles R, et al: Red cell fragmentation (schis- tions and pathophysiologic mechanisms. Urology 1986;27:389.
tocytosis) after bone marrow transplantation. Bone Marrow 270. Bergstein J, Andreoli SP, Provisor AJ, Yum M: Radiation
Transplant 1998;22:777–780. nephritis following total-body irradiation and cyclophos-
250. Fuge R, Bird JM, Fraser A, et al: The clinical features, risk phamide in preparation for bone marrow transplantation.
factors and outcome of thrombotic thrombocytopenic Transplantation 1986;41:63–66.
purpura occurring after bone marrow transplantation. Br J 271. Galli FC, Damon LE, Tomlanovich SJ, Keith F, Chatterjee K,
Haematol 2001;113:58–64. DeMarco T: Cyclosporine-induced hemolytic uremic syn-
251. Iacopino P, Pucci G, Arcese W, et al: Severe thrombotic drome in a heart transplant recipient. J Heart Lung Transplant
microangiopathy: An infrequent complication of bone 1993;12:440–444.
marrow transplantation. Gruppo Italiano Trapianto Midollo 272. Holman MJ, Gonwa TA, Cooper B, et al: FK506–associated
Osseo (GITMO). Bone Marrow Transplant 1999;24:47–51. thrombotic thrombocytopenic purpura. Transplantation
252. Pettitt AR, Clark RE: Thrombotic microangiopathy following 1993;55:205–206.
bone marrow transplantation. Bone Marrow Transplant 273. Gharpure VS, Devine SM, Holland HK, Geller RB, O’Toole K,
1994;14:495–504. Wingard JR: Thrombotic thrombocytopenic purpura associ-
514 Immune Hemolytic Anemias

ated with FK506 following bone marrow transplantation. 295. Guinan EC, Tarbell NJ, Niemeyer CM, Sallan SE, Seinstein HJ:
Bone Marrow Transplant 1995;16:715–716. Intravascular hemolysis and renal insufficiency after bone
274. Juckett M, Perry EH, Daniels BS, Weisdorf DJ: Hemolytic marrow transplantation. Blood 1988;72:451–455.
uremic syndrome following bone marrow transplantation. 296. Dzik W, Georgi BA, Khettry U, Jenkins RL: Cyclosporine-asso-
Bone Marrow Transplant 1991;7:405–409. ciated thrombotic thrombocytopenic purpura following liver
275. Marshall RJ, Sweny P: Haemolytic-uraemic syndrome in transplantation—successful treatment with plasma exchange.
recipients of bone marrow transplants not treated with Transplantation 1987;44:570–572.
cyclosporin A. Histopathology 1986;10:953–962. 297. Montaguti P, Melloni E, Cavalletti E: Acute intravenous toxic-
276. Chavers BM, Wells TG, Burke BA, Mauer SM: De novo ity of dimethyl sulfoxide, polyethylene glycol 400, dimethyl-
hemolytic uremic syndrome following renal transplantation. formamide, absolute ethanol, and benzyl alcohol in inbred
Ped Nephrol 1990;4:62–64. mouse strains. Arzneim-Forsch Drug Res 1994;44:566.
277. Johnson PRE, Yin JA, Drayson MT: Thrombotic thrombocy- 298. Samoszuk M, Reid ME, Toy PTCY: Intravenous dimethylsul-
topenic purpura following allogeneic bone marrow transplan- foxide therapy causes severe hemolysis mimicking a
tation [letter]. Bone Marrow Transplant 1991;7:321–325. hemolytic transfusion reaction. Transfusion 1983;23:405.
278. Kwaan HC: Miscellaneous secondary thrombotic microan- 299. DiStefano V, Kahn JJ: Observations on the pharmacology and
giopathy [review]. Semin Hematol 1987;24:141–147. hemolytic activity of dimethylsulfoxide. Toxicol Appl
279. Van Buren D, Van Buren CT, Lechner SM, Addox AM, Erani R, Pharmacol 1965;7:660–666.
Ahan BD: De novo hemolytic uremic syndrome in renal trans- 300. Smith DM, Weisenburger DD, Bierman P, Kessinger A,
plant recipients immunosuppressed with cyclosporine. Vaughan WP, Armitage JO: Acute renal failure associated with
Surgery 1985;98:54–62. autologous bone marrow transplantation. Bone Marrow
280. Buturovic J, Kandus A, Malovrh M, Bren A, Drinovec J: Transplant 1987;2:195–201.
Cyclosporine-associated hemolytic uremic syndrome in four 301. Burger J, Gilmore MJ, Jackson B, Prentice HG: Acute haemo-
renal allograft recipients: Resolution without specific therapy. globinaemia associated with the reinfusion of bone marrow
Transplant Proc 1990;22:1726–1727. buffy coat for autologous bone marrow transplantation
281. Schwarz A, Krause PH, Offermann G, Keller F: Recurrent and [letter]. Bone Marrow Transplant 1991;7:322–324.
de novo renal disease after kidney transplantation with or 302. Kessinger A, Schmit Pokorny K, Smith D, Armitage J:
without cyclosporine A. Am J Kid Dis 1991;17:524–531. Cryopreservation and infusion of autologous peripheral
282. Dzik WH, Jenkins RL: Renal failure from ABO hemolysis due blood stem cells. Bone Marrow Transplant 1990;5(Suppl.
to anti-A of graft origin following liver transplantation. 1):25–27.
Transfusion 1987;27:550. 303. Chaplin H: Abdominal pain, total intravascular hemolysis, and
283. Bonser RS, Adu D, Franklin I, McMaster P: Cyclosporin- death in a 53-year-old woman. Am J Med 1990;88:667–674.
induced haemolytic uraemic syndrome in liver allograft recip- 304. Becker RC, Giuliani M, Savage RA, Weick JK: Massive hemo-
ient [letter]. Lancet 1984;ii:1337. lysis in Clostridium perfringens infections. J Surg Oncol
284. Gross M, Hiesse C, Kriaa F, Goldwasser F: Severe hemolytic 1987;35:13–18.
uremic syndrome in an advanced ovarian cancer patient 305. Terebelo HR, McCue RL, Lenneville MS: Implication of
treated with carboplatin and gemcitabine. Anticancer Drugs plasma free hemoglobin in massive clostridial hemolysis.
1999;10:533–536. JAMA 1982;248:2028–2029.
285. Nordstrom B, Strang P: Microangiopathic hemolytic anemias 306. Hubl W, Mostbeck B, Hartleb H, Pointner H, Kofler K, Bayer
(MAHA) in cancer: A case report and review [review]. PM: Investigation of the pathogenesis of massive hemolysis in
Anticancer Res 1993;13:1845–1850. a case of Clostridium perfringens septicemia. Ann Hematol
286. Jackson AM, Rose BD, Graff LG, et al: Thrombotic micro- 1993;67:145–147.
angiopathy and renal failure associated with antineoplastic 307. Pun KC, Wehner JH: Abdominal pain and massive intravas-
chemotherapy. Ann Intern Med 1984;101:41–44. cular hemolysis in a 47–year-old man. Chest 1996;110:
287. Gordon LI, Kwaan HC: Cancer- and drug-associated throm- 1353–1355.
botic thrombocytopenic purpura and hemolytic uremic syn- 308. Ifthikaruddin JJ, Holmes JA: Clostridium perfringens septi-
drome. Semin Hematol 1997;34:140–147. caemia and massive intravascular haemolysis as a terminal
288. Bennett CL, Weinberg PD, Rozenberg-Ben-Dror K, Yarnold complication of autologous bone marrow transplant. Clin Lab
PR, Kwaan HC, Green D: Thrombotic thrombocytopenic Haematol 1992;14:159–161.
purpura associated with ticlopidine: A review of 60 cases. Ann 309. Eschbach JW, Korn D, Finch CA: 14C cyanate as a tag for red
Intern Med 1998;128:541–544. cell survival in normal and uremic man. J Lab Clin Med
289. Chen DK, Kim JS, Sutton DM: Thrombotic thrombocytopenic 1977;89:823–828.
purpura associated with ticlopidine use: A report of 3 cases 310. Korzets A, Ori Y, Grinblat J, Sirkis Y, Zevin D: Acute intravas-
and review of the literature. Arch Intern Med 1999;159: cular haemolysis associated with an indwelling haemodialy-
311–314. sis catheter [letter]. Transplantation 1990;5:1055–1057.
290. Gottschall JL, Elliot W, Lianos E, McFarland JG , Wolfmeyer K, 311. Bird GWG, Wingham J: Anti A autoantibodies with unusual
Aster RH: Quinine-induced immune thrombocytopenia asso- properties in a patient on renal dialysis. Immunol Commun
ciated with hemolytic uremic syndrome: A new clinical entity. 1980;9:155.
Blood 1991;77:306–310. 312. Lundgren G, Asaba H, Bergstrom J, et al: Fulminating anti-A
291. Gottschall JL, Neahring B, McFarland JG, et al: Quinine- autoimmune hemolysis with anuria in a renal transplant
induced immune thrombocytopenia with hemolytic uremic recipient: A therapeutic role of plasma exchange. Clin
syndrome: Clinical and serological findings in nine patients Nephrol 1981;16:211–214.
and review of literature. Am J Hematol 1994;47:283–289. 313. Mangal AK, Growe GH, Sinclair M, Stillwell GF, Reeve CE,
292. Stroncek DF, Vercellotti GM, Hammerschmidt DE, Christie DJ, Naiman SC: Acquired hemolytic anemia due to “auto”-anti-A
Shankar RA, Jacob HS: Characterization of multiple quinine- or “auto”-anti-B induced by group O homograft in renal
dependent antibodies in a patient with episodic hemolytic transplant recipients. Transfusion 1984;24:201–205.
uremic syndrome and immune agranulocytosis. Blood 314. Nyberg G, Sandberg L, Rydberg L, et al: ABO-autoimmune
1992;80:241–248. hemolytic anemia in a renal transplant patient treated with
293. Aster RH: Quinine sensitivity: A new cause of the hemolytic cyclosporin: A case report. Transplantation 1984;37:529–530.
uremic syndrome. Ann Intern Med 1993;119:243–244. 315. Povlsen JV, Rasmussen A, Hansen HE, Fjeldborg O,
294. Maguire RB, Stroncek DF, Campbell AC: Recurrent pancy- Kissmeyer-Nielsen F, Madsen M: Acquired haemolytic
topenia, coagulopathy, and renal failure associated with mul- anaemia due to isohaemagglutinins of donor origin following
tiple quinine-dependent antibodies [review]. Ann Intern Med ABO-minor-incompatible kidney transplantation. Nephrol
1993;119:215–217. Dial Transplant 1990;5:148–151.
Immune Hemolysis Associated with Transplantation 515

316. Herron R, Clark M, Tate D, Kruger A, Smith DS: Immune bodies after minor ABO-mismatched heart and lung trans-
haemolysis in a renal transplant recipient due to antibodies plantation. Transplantation 1988;46:246–249.
with anti-c specificity. Vox Sang 1986;51:226–227. 339. Knoop C, Adrien M, Antoine M, et al: Severe hemolysis due to
317. Ramsey G, Israel L, Lindsay GD, Mayer TK, Nusbacher J: a donor anti-D antibody after heart-lung transplantation:
Anti-Rho(D) in two Rh-positive patients receiving kidney Association with lung and blood chimerism. Am Rev Respir
grafts from an Rh-immunized donor. Transplant Dis 1993;148:504–506.
1986;41:67–69. 340. Mazzara R, Pereira A, Vilardell J, Andreu J, Ricart MJ, Castillo
318. Watzke H, Kovarik J, Gassner H: A permissive effect of R: ‘AUTO’-anti-ABO antibodies after organ transplantation: A
cyclosporin on the development of isohaemagglutinins of Ciclosporin A related phenomenon? [letter]. Vox Sang
graft origin in ABO-mismatched organ transplantation. 1988;55:246.
Nephrol Dial Transplant 1988;3:89–90. 341. Salamon DJ, Ramsey G, Nusbacher J, Yang S, Starzl TE, Israel
319. Gregoire JR: Immune hemolytic anemia after renal transplan- L: Anti-A production by a group O spleen transplanted to a
tation secondary to ABO- minor-mismatch between the donor group A recipient. Vox Sang 1985;48:309–312.
and recipient. J Am Soc Nephrol 1993;4:1122–1126. 342. Ramsey G, Sherman LA: Transfusion therapy in solid organ
320. Borka P, Jakab J, Rajczy K, et al: Temporary donor-derived B- transplantation. Hematol Oncol Clin North Am 1994;8:
lymphocyte microchimerism leading to hemolysis in minor 1117–1129.
ABO-incompatible renal transplantation. Transplant Proc 343. Swanson J, Sebring E, Sastamoinen R, Chopek M: Gm allo-
2001;33:2287–2289. typing to determine the origin of the anti-D causing hemolytic
321. Peces R, Navascues RA, Laures AS, et al: Alloimmune anemia in a kidney transplant recipient. Vox Sang 1987;
haemolytic anaemia resulting from anti-A and anti-B antibody 52:228–230.
induced by group O graft in renal transplant recipients. Nephrol 344. Swanson JL, Sastamoinen RM, Steeper TA, Sebring ES: Gm
Dial Transplant 1998;13:1866–1869. allotyping to determine the origin of red cell antibodies in
322. Bertoni E, Rosati A, Zanazzi M, et al: Graft-versus-host anti- recipients of solid organ transplants. Vox Sang 1987;52:75–78.
body reactions in ABO unmatched renal transplants. 345. Ahmed KY, Nunn G, Brazier DM, Bird GW, Crockett RE:
Transplant Proc 1998;30:1333–1334. Hemolytic anemia resulting from autoantibodies produced by
323. Frohn C, Jabs WJ, Fricke L, Goerg S: Hemolytic anemia after the donor’s lymphocytes after renal transplantation.
kidney transplantation: Case report and differential diagnosis. Transplantation 1987;43:163–164.
Ann Hematol 2002;81:158–160. 346. Orchard J, Young NT, Smith C, Thomas S, Darke C: Severe
324. Odabas AR, Tutucu KN, Turkmen A, Keskin H, Sever MS: intravascular haemolysis in a renal transplant recipient due to
Severe alloimmune hemolytic anemia after renal transplanta- anti-B of donor origin. Vox Sang 1990;59:172–175.
tion. Nephron 2002;92:743–745. 347. Brecher ME, Moore SB, Reisner RK, Rakela J, Krom RAF:
325. Ramsey G, Nusbacher J, Starzl TE, Lindsay GD: Isohe- Delayed hemolysis resulting from anti-A1 after liver trans-
magglutinins of graft origin after ABO-unmatched liver trans- plantation. Am J Clin Pathol 1989;91:232–235.
plantation. N Engl J Med 1984;311:1167–1170. 348. Swanson J, Sastamoinen R, Sebring E, Chopek M: Rh and Kell
326. Ramsey G, Cornell FW, Hahn L, et al: Red cell antibody alloantibodies of probable donor origin produced after solid
problems in 1000 liver transplants [abstract]. Transfusion organ transplantation. Transfusion 1985;25:467.
1987;27:552. 349. Seltsam A, Hell A, Heymann G, Salama A: Donor-derived
327. Triulzi DJ, Shirey RS, Ness PM, Klein AS: Immunohema- alloantibodies and passenger lymphocyte syndrome in two of
tologic complications of ABO-unmatched liver transplants. four patients who received different organs from the same
Transfusion 1992;32:829–833. donor. Transfusion 2001;41:365–370.
328. Lee JH, Mintz PD: Graft versus host anti-Rho(D) following 350. Bradley R, Triulzi DJ, Starzl TE: Donor derived red cell anti-
minor Rh-incompatible orthotopic liver transplantation. Am J bodies in liver transplant recipients treated with FK-506.
Hematol 1993;44:168–171. Transfusion 1993;33:43S.
329. Hareuveni M, Merchav H, Austerlitz N, Rahimi-Levene N, 351. Jenkins RL, Georgi BA, Gallik-Karlson CA, et al: ABO mis-
Ben Tal O: Donor anti-Jk(a) causing hemolysis in a liver trans- match and liver transplantation. Transplant Proc 1987;19:
plant recipient. Transfusion 2002;42:363–367. 4580–4585.
330. Au WY, Lo CM, Fan ST, Liu CL, Lam CC: Life-threatening 352. Jacobs LB, Shirey RS, Ness PM: Hemolysis due to the simulta-
ABO-mediated hemolysis after cadaveric orthotopic liver neous occurrence of passenger lymphocyte syndrome and a
transplantation. Transplantation 2002;74:285–286. delayed hemolytic transfusion reaction in a liver transplant
331. Bird GW, Wingham J: Formation of blood group “autoanti- patient. Arch Pathol Lab Med 1996;120:684–686.
bodies” after transplantation. Transfusion 1982;22:400. 353. Bracey AW, Van Buren C: Anti-A donor lymphocytic origin in
332. Marchioro TL, Rowlands DT, Rifkind D, et al: Splenic homo- three recipients of organs from the same donor. Vox Sang
transplantation. Ann NY Acad Sci 1964;120:626. 1987;53:181–183.
333. Taaning E, Morling N, Mortensen SA, Pettersson G, Simonsen 354. Bapat AR: Thrombocytopenia and autoimmune hemolytic
AC: Severe hemolysis caused by graft-derived anti-B produc- anemia following renal transplantation. Transplantation
tion after lung transplantation. J Heart Lung Transplant 1987;44:157.
1995;15:850–851. 355. Larrea L, de la Rubia J, Arriaga F, Sanchez J, Marty ML: Severe
334. Salerno CT, Burdine J, Perry EH, Kshettry VR, Hertz MI, hemolytic anemia due to anti-E after renal transplantation.
Bolman RM III: Donor-derived antibodies and hemolysis after Transplantation 1997;64:550–551.
ABO-compatible but nonidentical heart-lung and lung trans- 356. Petz LD, Calhoun L: Preparation for blood and blood product
plantation. Transplantation 1998;65:261–264. replacement. In Busuttil RW, Klintmalm GB (ed):
335. Solheim BG, Albrechtsen DA, Berg KJ, et al: Hemolytic Transplantation of the Liver. Philadelphia: WB Saunders,
anemia in cyclosporine-treated recipients of kidney or heart 1996:442–448.
grafts from donors with minor incompatability for ABO anti- 357. Minakuchi J, Toma H, Takahashi K, Ota K: Autoanti-A and -B
gens. Transplant Proc 1987;19:4236–4238. antibody induced by ABO unmatched blood group kidney
336. Solheim BG, Albrechtsen DA, Egeland T, et al: Auto-antibodies allograft. Transplant Proc 1985;17:2297–2300.
against erythrocytes in transplant patients produced by donor 358. Ota K, Takahashi K, Toma H: Cyclosporine-treated kidney
lymphocytes. Transplant Proc 1987;19:4520–4521. transplants from living-related donors—a single center trial.
337. Albrechtsen DA, Solheim BG, Flatmark A, et al: Autoimmune In Terasaki P (ed): Clinical Transplants. Los Angeles: UCLA
hemolytic anemia in cyclosporine-treated organ allograft Tissue Typing Laboratory, 1988:189–198.
recipients. Transplant Proc 1988;20:959–962. 359. Ishikura H, Endo J, Saito Y, et al: Graft-versus-host antibody
338. Hunt BJ, Yacoub M, Amin S, Devenish A, Contreras M: reaction causing a delayed hemolytic anemia after blood
Induction of red blood cell destruction by graft-derived anti- transfusion [letter]. Blood 1993;82:3222–3223.
516 Immune Hemolytic Anemias

359a. Petz LD, Garratty G: Acquired Immune Hemolytic Anemias, 373. Ramsey G, Hahn LF, Cornell FW, et al: Low rate of Rhesus
1st ed. New York: Churchill-Livingstone, 1980:331–337. immunization from Rh-incompatible blood transfusions
360. Salama A, Mueller-Eckhardt C: Delayed hemolytic transfusion during liver and heart transplant surgery. Transplantation
reactions: Evidence for complement activation involving allo- 1989;47:993–995.
geneic and autologous red cells. Transfusion 1984;24:188–193. 374. Blomqvist BI, Wikman A, Shanwell A, Eleborg L: Erythrocyte
361. Ness PM, Shirey RS, Thoman SK, Buck SA: The differentiation antibodies in liver transplantation: Experiences from Huddinge
of delayed serologic and delayed hemolytic transfusion reac- University Hospital. Transplant Proc 1991;23:1944–1945.
tions: Incidence, long-term serologic findings, and clinical 375. Forsyth C, Kronenberg H, McCaughan G: Severe hemolysis
significance. Transfusion 1990;30:688–693. due to anti-D in D-negative recipient of an orthotopic liver
362. Schechter GP, Whang-Peng J, McFarland W: Circulation of transplant. Transfusion 1995;35:277–278.
donor lymphocytes after blood transfusion in man. Blood 376. Hyma BA, Moore SB, Grande JP, et al: Delayed immune
1977;49:651–656. hemolysis in a patient receiving cyclosporine after orthotopic
363. Adams PT, Davenport RD, Reardon DA, Roth MS: Detection liver transplantation. Transfusion 1988;28:276–279.
of circulating donor white blood cells in patients receiving 377. Casanueva M, Valdes MD, Ribera MC: Lack of alloimmuniza-
multiple transfusions. Blood 1992;80:551–555. tion to D antigen in D-negative immunosuppresed liver trans-
364. Lee TH, Donegan E, Slichter S, Busch MP: In vivo prolifera- plant recipients. Transfusion 1994;34:570–572.
tion of donor leukocytes following allogeneic transfusions of 378. Rougier JP, Viron B, Ronco P, Khayat R, Michel C, Mignon F:
immunocompetent recipients. Transfusion 1993;33:51S. Autoimmune haemolytic anemia after ABO-match, ABDR full
365. Hutchinson RM, Sejeny SA, Fraser ID, Tovey GH: match kidney transplantation. Nephrol Dial Transplant
Lymphocyte response to blood transfusion in man: A compar- 1994;9:693–697.
ison of different preparations of blood. Br J Haematol 379. McMillan MA, Muirhead CSM, Lucie NP, Briggs JD, Junor BJ:
1976;33:105–111. Autoimmune haemolytic anaemia related to cyclosporine
366. Dzik WH: Mononuclear cell microchimerism and the immuno- with ABO-compatible kidney donor and recipient. Nephrol
modulatory effect of transfusion. Transfusion 1994;34:1007. Dial Transplant 1991;6:57–59.
367. Starzl TE, Demetris AJ, Trucco M, et al: Chimerism after liver 380. De Vecchi A, Zanella A, Egidi F, Ponticelli C: Autoimmune
transplantation for type IV glycogen storage disease and type hemolytic anemia in a cadaveric renal transplant recipient
1 Gaucher’s disease. N Engl J Med 1993;328:745–749. treated with cyclosporine. Acta Haematol 1985;73:216–218.
368. Fung JJ: Tolerance and chimerism in liver transplantation. 381. Beiting CY, Larimore KS: A case report: Cold hemagglutinin
Transplant Proc 1997;29:2817–2818. disease in a pancreatic and renal transplant patient.
369. Brodthagen UA, Bud M: Rhesus immunization after Rh- Immunohematology 1988;4:85–87.
incompatible kidney transplantation. Tissue Antigens 382. West KA, Anderson DR, McAlister VC, et al: Alloimmune
1986;27:102–105. thrombocytopenia after organ transplantation. N Engl J Med
370. Huestis DW, Zukoski CF: Secondary stimulation of Rh anti- 1999;341:1504–1507.
bodies by kidney allografts from Rh-positive donors. Vox 383. Friend PJ, McCarthy LJ, Filo RS, et al: Transmission of idio-
Sang 1973;24:524–527. pathic (autoimmune) thrombocytopenic purpura by liver
371. Kenwright MG, Sangster JM, Sachs JA: Development of transplantation. N Engl J Med 1990;323:807–811.
RhD antibodies after kidney transplantation. Br Med J 384. Jondeau G, Bierling P, Rossert J, et al: Autoimmune neutrope-
1976;2:151–152. nia after renal transplantation. Transplantation 1988;46:589.
372. Gluckman JC, Foucault C, Beaufils H, et al: Rh antibodies after 385. Wood K, Sachs D: Chimerism and transplantation tolerance:
kidney transplantation. Transplantation 1981;32:260–262. Cause and effect. Immunol Today 1996;17:584
C H A P T E R 1 3

Hemolytic Disease of the


Fetus and Newborn

Hemolytic disease of the fetus Unfortunately, such reports are often perpetuated
and newborn (HDFN) is a secondhand by authors quoting the original reference
condition in which fetal or (often without critically reading the original report),
newborn infants’ RBCs have a or the antibody is added to a list of antibodies pur-
shortened life span due to ported to cause HDFN. When we refer to HDFN in
maternal antibodies. These this chapter, it implies that a hemolytic anemia was
antibodies cross the placenta present in the fetus and/or the newborn infant, and
and sensitize the fetal RBCs; the antibodies we list as a cause of HDFN are only
they are usually IgG alloanti- those for which there is evidence of having caused an
bodies but on rare occasions immune hemolytic anemia.
can be IgG maternal autoantibodies. This condition is The first antibody to be described as a cause of
more commonly called hemolytic disease of the HDFN was anti-D. Until the use of Rh immunopro-
newborn (HDN), but we (and others) prefer the more phylaxis in 1968, anti-D was, by far, the most
accurate term, HDFN.1,2 The hemolytic process varies common antibody to cause HDFN, being responsible
from severe in utero hemolysis early in pregnancy, for about 98% of all cases.2,3 Approximately one in
resulting in fetal death (often associated with hydrops 180 of all newborn white infants suffered with HDFN
fetalis), to a mild process that might not be noticeable due to anti-D.2 In 1964, Giblett3 reported that 93% of
until a day or more after the baby’s birth. Sensitization antibodies detected in sera of pregnant women were
of the baby’s RBCs with maternal antibody does not anti-D (or anti-C+D); 6% were of other Rh speci-
necessarily lead to HDFN. Many babies are born with ficities. In 1967, Polesky4 reported that 5.7% of preg-
a strongly positive direct antiglobulin test (DAT) due nant women were alloimmunized; 83% of these
to IgG (even Rh antibody) sensitization but have no involved Rh antibodies, and 1% were associated with
obvious signs of a hemolytic anemia. This phenome- specificities other than Rh. In 1969, Queenan and col-
non has led to some confusion in the literature; in leagues5 reported that 3.1% of pregnant women were
many reports, antibodies are described as being a alloimmunized; 68% of these were associated with
cause of HDFN because the baby was born with a pos- anti-D and 1.3% with specificities other than Rh.
itive DAT due to a specific maternal antibody, and yet Since Rh prophylaxis became widespread, the picture
no (or very little) evidence has been presented for the has changed, and anti-D is much less common. The
presence of hemolytic anemia. This is particularly the comparative incidence of alloantibodies other than
case for ABO HDFN (see the later discussion). anti-D has also been affected by the increased
517
518 Immune Hemolytic Anemias

population of pregnant group women who have been patients are becoming increasingly more commonly
transfused; Queenan and colleagues5 reported that a immunized to antigens other than D, anti-D is still
history of prior transfusion was nine times more fre- probably the most common cause of HDFN (i.e., it
quent among alloimmunized pregnant women. There might not be the most commonly detected antibody,
are a few reports available to illustrate the changing but it still has the greatest potential to destroy fetal
picture after the introduction of Rh prophylaxis. RBCs). Although almost any IgG antibody must be
Walker6 reported that in Michigan (US) in 1969, one considered to have the potential to cause HDFN, very
in every 175 (0.6%) obstetric patients produced anti- few other than anti-D appear to cause HDFN severe
D, but that by 1984 this rate had fallen to 1.7 per 1000 enough to require transfusion. There are many factors
(0.17%) (1.4% of Rh[D] negative women with Rh[D] that affect the clinical significance of maternal allo-
positive husbands). Over a 12-year period at Walker’s antibodies (Table 13-2). Sometimes the respective
hospital, 104 of 55,877 (1/537) deliveries were associ- antigen is not very well developed on the fetal RBCs
ated with antibodies of potential clinical significance (Table 13-3); thus, even though a strong IgG antibody
other than anti-D. Historically, the next most common is present in the mother’s serum, the antibody might
antibody after anti-D was anti-E (1/1693 or 0.06%), cause no HDFN or only a mild case.
followed by anti-c, -Jka, and -K (approximately Anti-c is usually described as being the next most
1/4000 or 0.3%); these were followed by anti-C, -s, -e, common cause of severe HDFN after anti-D.
-cE, -Fya (approximately 1/14,000 or 0.007%).6 Nevertheless, Mollison and coworkers2 report that
Kornstad7 reported that in Norway, the occurrence of only about 30% of babies born with RBCs sensitized
new cases of anti-D had fallen from 57.6 cases per with anti-c require exchange transfusion, in contrast
10,000 (0.6%) women in 1967–1969 to 16.5 per 10,000 to 60% of babies born with anti-D–sensitized red cells.
(0.2%) women during the period 1981–1983. Fifty- Wenk and associates10 reported that the frequency of
eight percent of the new cases of immunization to Rh anti-c was 0.52 that of anti-D in their obstetric
antigens occurred in Rh(D)-negative women and 42% patients. Of 70 cases with maternal anti-c and c-
in Rh(D)-positive women. Rh antibodies other than positive babies, 16 babies (23%) had negative DATs
anti-D accounted for 53% and anti-D for 47% of the and no evidence of HDFN. Of 46 cases who had
“new” antibodies. In an earlier report, Kornstad8 HDFN, eight (17%) had mild disease and required no
reported on antibodies that were detected in 148,200 transfusion, 20 (43%) had moderate disease and
Rh(D)-positive women during a five-year period required transfusion, and 8 (17%) died. Sixty-seven
(1975–1980). These included 99 antibodies with Rh percent of the c-alloimmunized women who deli-
specificities other than anti-D, and 101 antibodies of vered babies with HDFN had a history of prior blood
specificities other than Rh that had the potential to transfusion; there was a higher incidence of transfu-
cause HDFN. Anti-E was the most commonly sion in the group of mothers delivering severely
detected antibody in D-positive women, followed affected babies. Although anti-K is probably the most
by anti-K, -c, -Cw, and -Fya. In the same period, clinically potent antibody after ABO and Rh, it rarely
204 D-negative women had anti-D detected in their causes HDFN, as only 9% of the white population (i.e.,
sera. fathers of babies) are K positive. In two studies of
Between 12% and 18% of white women are D 477,000 pregnancies, 0.1% of the women’s sera
negative, but a much lower frequency is found in contained anti-K; 78% of these were transfusion-
other populations. The D-negative phenotype is rare induced.11,12 As most fathers are K heterozygotes (Kk),
(1.7%) among Chinese, Japanese, and Southeast fewer than 5% of K-negative mothers will carry a K-
Asians and occurs in only 2%–5% of Africans. Some of positive baby. Table 13-4 shows data from six publica-
the Asian D-negative population have a weak D tions on HDFN associated with anti-K.13-18
antigen (Del) and do not make anti-D. Thus, HDFN The pathogenesis of anti-K–related HDFN is often
due to anti-D is rare among nonwhites. different from Rh disease. It was noted some years ago
that babies could be born with severe HDFN due to
anti-K and yet have negative DATs and/or low ΔOD
ANTIBODIES OF POTENTIAL values.19,20 Vaughan and colleagues21,22 showed that
CLINICAL SIGNIFICANCE PRODUCED unlike Rh antibodies, anti-K can react with erythroid
DURING PREGNANCY precursor cells. Thus, one component of the fetal/
newborn anemia associated with anti-K could be due
Antibodies That Cause HDFN to suppressed erythropoiesis; extravascular hemolysis
most often is present also.
Approximately 3%–6% of pregnant women have Hardy and Napier23 reviewed cases of HDFN in
antibodies (other than ABO) of potential clinical Wales (1948–1978) that were associated with antibodies
significance.2-9 Only antibodies that can cross the pla- other than anti-D. Of 557 women with Rh antibodies
centa cause HDFN; this restricts the cause of clinical other than anti-D, 388 delivered babies whose RBCs
problems to IgG antibodies. Table 13-1 shows anti- had the relevant antigen, but only 75% of these had a
bodies, other than ABO, that have been reported to positive DAT, and only 15% required treatment. There
cause HDFN. Although it is clear that obstetric were 128 women with non-Rh antibodies (e.g., anti-K,
Hemolytic Disease of the Fetus and Newborn 519

TABLE 13-1. BLOOD GROUP ANTIBODIES OTHER THAN ABO REPORTED AS A CAUSE OF HDFN

Blood Group Severity of HDFN Most Blood Group Severity of HDFN Most
System/Collection Antibody Often Encountereda,b System/Collection Antibody Often Encountereda,b

Rh D Moderate-severe Mv Mild
C Moderate-severe MiIII Severe
E Moderate-severe Mit Mild
c Moderate-severe Mta Mild-severe
e Mild-severe Vw Mild-severe
Cw Mild-moderate Mur Mild
Cx Mild-severe Hil Mild
Ew Moderate-severe Hut Mild
G Moderate Ena Severe
Goa Severe Globoside P Mild
f(ce) Severe
Ce Severe Lutheran Lua Mild
Rh:14 Moderate-severe Lub Mild
Rh:15 Moderate-severe Lu9 Mild
Rh:16 Moderate-severe Diego Dia Mild-severe
Rh:29 Moderate-severe Dib Mild-severe
Rh:32 Moderate-severe Wra Mild-severe
Rh:36 Moderate-severe ELO Mild-severe
Rh:37 Moderate-severe Colton Coa Mild-severe
Bea ? Cob Severe
Evans ? Co3 Severe
Tar ?
Rh:42 Mild Gerbich Ge Mild-severe
Riv Mild Landsteiner-Weiner LW Mild-moderate
Jal Mild High-Frequency Antigens
Stem Mild Augustine Ata Mild
Kell K Mild-severe Junior Jra Mild
k Mild-moderate Langereis Lan Mild-moderate
Kpa Mild Vel Vel Mild
Kpb Mild-severe Low-Frequency Antigens
Jsa Moderate Biles Bi Mild
Jsb Mild-severe Batty By Mild
Ku Mild-severe Froese Fra Mild
Ula Mild-severe Kamhuber Far(Kam) Severe
K22 Mild-severe Livesey Mild
Duffy Fya Mild-severe Gambino Ga Mild
Fyb Moderate Good Severe
Heibel Severe
Kidd Jka Mild-severe
Hta Hta Mild
Jkb Mild
Radin Rd Mild
Jk3 Mild
Reid Rea
MNSs M Mild-severe Zd Zd Severe
N Mild Jones/Hol Mild-moderate
‘N’ Moderate Scianna Sc2 Moderate
S Severe Gonzales Goa Mild
s Severe HJK HJK Severe
U Severe Kg Kg Severe
Mia Mild-severe REIT REIT Severe
a Mild = no transfusions needed; phototherapy usually used
Moderate = transfusion given, but sometimes only after delivery
Severe = fetal death IUT and/or exchange transfusion
b Most examples of the antibodies listed, except anti-D, cause no HDFN. Severity relates to cases reported to have evidence of hemolytic anemia; this is sometimes a
single case report.

–Fy, –Jk, etc.). These women delivered 35 babies whose had bilirubin levels exceeding 12.9 mg/dL. In only one
RBCs had the appropriate antigen; 91% of these had a case (anti-E) was an exchange transfusion required; this
positive DAT, but only 6% (one anti-Fya and one anti- was 40 hours after birth and perhaps not related to the
Kpa) required treatment. Walker6 reviewed his experi- anti-E sensitization.
ences over a 12-year period (1972–1983) in which 104 In Manitoba, Canada (population 1 million), the
examples of IgG antibodies other than anti-D were mean annual occurrence of D alloimmunization in
encountered; this represented one in 537 deliveries. pregnant women dropped from 194 in the 5-year
Approximately 71% of these infants were unaffected by period ending October 31, 1967 to 28 in the 6-year
the antibodies; 22% had early onset of jaundice, and 7% period ending October 31, 1988.11,13 In the same two
520 Immune Hemolytic Anemias

periods, the mean annual occurrence of alloimmu-


TABLE 13-2. FACTORS THAT COULD AFFECT nization associated with alloantibodies other than
SIGNIfiCANCE OF MATERNAL ALLOANTIBODIES anti-D increased from 14 to 88. It was suggested that
this increase was due partially to the increased
1. Class and subclass of antibody screening of pregnant D-positive women. This
2. Strength/quantity of antibody
3. Presence, or strength, of antigen on fetal RBCs (see Table 13-3) increase was also likely due to an increase in the fre-
4. Efficiency of placental transfer quency of blood transfusion. It should be noted that
5. Efficiency of fetal reticuloendothelial system 48% of babies born to mothers with anti-D were
6. Competition effect of antigen present in fetal body fluids or affected (i.e., RBCs were sensitized with maternal
fetal tissue
7. Maternal blocking antibodies (capable of blocking fetal
antibody), but only 51% of these required treatment.
macrophage receptors) If one excludes the one case associated with anti-k,
2.4%–65% of babies born to mothers having antibo-
dies other than anti-D showed any signs of being
affected, and only 12%–50% of these required treat-
ment. Table 13-5 shows data, published by Bowman,
on the severity of HDFN associated with all antibo-
dies detected in a 26-year period.11,13 These data rep-
TABLE 13-3. PRESENCE AND RELATIVE STRENGTH resent experiences of 1442 cases of HDFN. Almost
OF ANTIGENS ON CORD CELLS half of D-positive babies born to mothers with anti-D
did not require therapy. Up to 88% of antigen-
Antigen Strength Blood Group System or Antigen positive babies born to mothers with other antibo-
dies (including Rh antibodies other than anti-D) did
Well developed at birth MNSsU not require therapy.
Rh The specificity of the antibody should be a good
Kell
Duffy clue to its potential to cause HDFN (see Table 13-1). In
Kidd some cases (e.g., anti-M), it could be useful to deter-
Diego mine the immunoglobulin class (IgG or IgM) of the
Dombrock antibody, which can usually be easily done using 2-
Scianna
Gerbich
mercaptoethanol (2ME) or dithiothreitol (DTT). If the
Ena antibody is IgG and has the potential for causing
Ytb HDFN, knowledge of the father’s genotype is another
Present at birth but weaker aid to predicting whether the baby is likely to be
than on adult red cells ABH affected by the antibody. When anti-D is involved, the
P chances are that the baby’s father will be D positive,
Lutheran
Xga
but it is useful to determine the father’s Rh phenotype
and hence the possible genotype. This information is
Very weak or absent at birth
Yta also useful if the infant is born affected and the
Vel mother wants to plan further pregnancies (i.e., the
Lewis child of a father homozygous for D will always be D
I positive, and the outcome of future pregnancies will
Sda
Chido
be similar or worse). Phenotyping the father’s RBCs is
especially useful when antibodies other than Rh are

TABLE 13-4. HDFN ASSOCIATED WITH ANTI-K

No. of Mothers No. of Affected No. of Babies


Reference Period Studied No. of Anti-K Previously Transfused (%) Babies Treated or IUFDb

13 1977–1983 211 71 7 4
14 1969–1984 127b 67 13 9
15 1980–1989 407c 88 10 5
16 1944–1990 459 NA 20 8
17 1959–1995 156 63 21 13
18 1984–1996 65 45 18 ?11d
a K+ baby and/or +DAT, with or without HDFN
b Intrauterine fetal death (IUFD) or treated with transfusion and/or phototherapy
c 0.1% of all pregnancies during this period
d Not clear in publication
NA, not available
Hemolytic Disease of the Fetus and Newborn 521

transfusion history who did not have antibodies


TABLE 13-5. SEVERITY OF HEMOLYTIC DISEASE detected at the first visit were tested again at
IN MANITOBA DURING 26 YEARS11,13 28 weeks; all women were retested at 32–36 weeks.
Sampling at 28 weeks alone did not detect all the anti-
Number of % of Affected Not Requiring bodies developing during pregnancy—fewer than
Alloantibody Women Babies* Treatment
50% had appeared by this time. Clinically affected
D (13 yrs) 420 48 49 infants—in particular those requiring exchange trans-
E 350 31 88 fusions—were found much more commonly among
c, cE 183 65 62 pregnancies in which antibodies were detected at the
C, Ce, Cw 108 32 79 first visit (before 26 weeks) than among those in which
K 337 2.4 75
Kpa 6 33 50 antibodies developed later; this was found to be true
k 1 100 for both anti-D and anti-c.24,25 Deaths due to anti-D
Fya 23 22 80 were entirely confined to mothers with antibodies
S 14 57 75 detected at the first visit. Based on their findings,
*Affected = any signs suggesting HDFN (e.g., positive DAT because babies’ Bowell and colleagues24 proposed that the practice of
RBCs possess putatitve antigen) screening all mothers before 26 weeks should con-
tinue, but only unsensitized Rh-negative and pre-
viously transfused mothers should be retested at
involved. For instance, anti-K is usually caused by 28 weeks and at 34–36 weeks; the remaining Rh-
previous transfusions, and there is a good chance that positive women need only be retested at 34–36 weeks.
the father and child will be K negative. It must be emphasized that if this later screening is
Table 13-1 lists antibodies associated with HDFN done, care must be taken not to misinterpret results
and the degree of severity reported. due to antenatal Rh immunoprophylaxis that could
There are many single-case reports in the literature have started at 28 weeks. The proposals suggested by
of other antibodies that are said to be associated with Bowell and colleages24 are in direct contrast to those of
HDFN, but no evidence is presented for a hemolytic Walker,6 who suggested (from a cost-benefit point of
process; often, the term HDFN is equated with view) that no screening is necessary for D-positive
the finding of antibodies in the mother and a baby women. Rothenberg and coworkers26 found that only
with a positive DAT. As has been emphasized by six of 9348 (0.06%) D-positive pregnant women devel-
Bowman’s11,13 data (see Table 13-5), a positive DAT oped new antibodies during the third trimester.
indicates that the fetal RBCs have become sensitized Heddle and associates27 found that 58 of 17,568
with antibody, but this does not equate with (0.24%) pregnant women had new antibodies detect-
hemolytic disease. able at delivery that were not present during first-
trimester testing. In both studies, no significant
neonatal problems were encountered due to these
Predicting the Presence and Severity new antibodies.
of HDFN The Scientific Section Coordinating Committee of
ANTIBODY SCREENING the American Association of Blood Banks (AABB) has
issued guidelines (not AABB standards) for serologi-
Following the discovery that anti-Rh(D) could cause cal testing of pregnant women.28 Table 13-6 shows
HDFN, it became common practice to screen the sera their recommendations for prenatal testing.
from all Rh(D)-negative pregnant women for Rh anti-
bodies. Later, when it was found that Rh(D)-positive
women could also have babies with HDFN due to Rh QUANTITATING THE AMOUNT
antibodies (other than anti-D) and non-Rh antibodies, OF SERUM ANTIBODY
it was suggested that sera from all pregnant women
be screened for antibodies. For some years, the most Until the advent of amniocentesis, antibody titers
common practice in the United States has been to were used routinely in an attempt to predict the sever-
screen sera from all women when they first present to ity of HDFN. After the advent of amniocentesis, indi-
the obstetrician. Sera from Rh-negative women are rect antiglobulin test (IAT) titers were usually used as
screened again at approximately 28 weeks, and some a guide to indicate the need to perform amniocentesis.
institutions routinely test pregnant women again at Titers provide only semiquantitative estimates and are
32–36 weeks. This later screening has received some not a measure of the total antibody concentration.
support from workers in Oxford, England. Bowell and Titers give only rough estimates of the amount of anti-
colleagues24 examined results of screening 70,000 sera body bound to the target RBCs and do not measure
from pregnant women. Irregular antibodies were the amount of antibody remaining free in solution at
found in 1% of the women, and two thirds of these the endpoint of agglutination. Hughes-Jones29 has
were found in D-positive women. Seventy percent of shown that the poor correlation between titer and
the antibodies were detectable in the sample taken at antibody concentration is dependent on the equili-
the first visit. Rh-negative women and women with a brium constant and heterogeneity index of the
522 Immune Hemolytic Anemias

TABLE 13-6. RECOMMENDED PRENATAL TESTING

Testing and Condition Timing

ABO
First pregnancy Initial visit
Subsequent pregnancies Initial visit
Other For pretransfusion testing
Rh (test for weak D optional)
First pregnancy Initial visit and at 26–28 weeks’ gestation
Subsequent pregnancies Initial visit
Other For pretransfusion testing
Unexpected Antibodies
All pregnancies Initial visit
D– pregnancies Before Rh Ig therapy (optional)
D+ pregnancies Third trimester if transfused or history of unexpected antibodies
Other For pretransfusion testing
Antibody Identification
Unexpected antibodies present Upon initial detection
Confirmatory testing At time of titration
Antibody Titration
Rh antibodies Upon initial detection
Repeat at 18–20 weeks’ gestation
Repeat at 2- to 4-week intervals if below critical titer (16–32)
Other potentially significant antibodies As above, with discussion with obstetrician

From Judd WJ: Practice guidelines for prenatal and perinatal immunotematology, revisited. Transfusion
2001;41:1445–1452.

antibody, the concentration of the RBCs, and the questioned. Most hospitals have a titer (e.g., 32) above
inherent error of the method. Complete reproduction which they recommend amniocentesis, but they rarely
is almost impossible, and only a difference in titer of have any data to support the use of this particular
at least two tubes (more than fourfold), or of a score of value; this has become known as the “critical antibody
10 (see Chapter 6), should be considered significant. titer” in the United States. Gall and Miller31 reported on
The coefficient of variation with manual titrations is a retrospective study of Rh-sensitized women over a
plus or minus 100% or more for intralaboratory repli- period of 24 years. Records of 202 obstetric patients
cates and much higher for interlaboratory estimates.30 representing 280 sensitized pregnancies from a pool of
Thus, it is perhaps not surprising that titers were not 39,910 deliveries were analyzed. A significant correla-
very good at predicting the severity of HDFN. tion between severity of HDFN and results of amniotic
Although an Rh-positive baby born to a mother who fluid analysis (ΔOD450), cord hematocrit, and bilirubin
has a high titer of anti-D (e.g., >100) is usually affected was noted. Although there was a significant correlation
to some degree, many severely affected babies have between antibody titer and severity of HDFN, 12
been born to mothers with relatively low titers (e.g., infants had mild to severe HDFN with titers of 16 or
8). Nevertheless, Bowman11,13 feels that titers have less, although it should be pointed out that only three
been very useful in his experience of monitoring of these babies needed treatment. Gall and Miller31 sug-
thousands of cases of HDFN over a period of more gested that critical antibody titer, as the initiating event
than 30 years. to begin amniocentesis, be abandoned. They suggested
When titers are performed routinely (e.g., at 2- to initiating amniocentesis on any woman who has
4-week intervals after 18 weeks gestation), the tech- detectable Rh antibody, regardless of the level.
nique should be standardized as much as possible (e.g., Nevertheless, in the United States, the “critical titer” is
same test system, same volume of sera/RBCs, RBC still in common use.
phenotype, and preferably the same technologist each In the United Kingdom, it is more common practice
time tests are performed), and it is very important that to quantitate anti-D in ng or IU/mL using an
the current serum sample be titrated in parallel with a AutoAnalyzer. Bowell and associates32,33 found, on
stored previous sample. A change (i.e., increase) in titer examination of the records of 288 pregnancies result-
under these conditions yields as much, if not more, ing in D-positive babies, that the chance of producing
valuable information as the absolute value. As men- a severely affected baby rises with increasing mater-
tioned previously, antibody titers are not usually used nal anti-D levels. In 140 pregnancies in which the
in current practice to predict the severity of HDFN but maternal anti-D level remained below 4 IU/mL, only
rather to indicate the need for amniocentesis and/or five babies required exchange transfusion, and no
other clinical studies. Because of the inaccuracies baby had a cord hemoglobin below 10 g/dL. On the
already mentioned, even the value of this should be other hand, of 148 pregnancies in which the anti-D
Hemolytic Disease of the Fetus and Newborn 523

level rose above 4 IU/mL, 99 babies required measured. Based on ΔOD450 measurements made after
exchange transfusions, and 25 babies had a cord 29 weeks in 101 Rh-sensitized pregnancies, Liley36
hemoglobin below 10 g/dL. The researchers con- divided his readings into three zones. He suggested
cluded that those pregnancies in which anti-D con- that fluids that gave ΔOD450 measurements in zone 3
centrations remained below 4 IU/mL represented a were indicative of severe HDFN (e.g., hydrops or fetal
safe group for whom amniocentesis should be death within 7 to 10 days); zone 1 indicated an unaf-
avoided. More recently, maternal anti-D concentra- fected or mildly affected baby, and zone 2 measure-
tions lower than 15 IU/mL were found to be associ- ments were intermediate. It was necessary to relate
ated with little or no fetal anemia as assessed by the ΔOD450 values to gestational age. There have been
fetoscopy.34 The extent of fetal hemolysis at high con- many reports confirming the value of this procedure
centrations was found to be highly variable. A cutoff to monitor the condition of the fetus. For instance,
value of 10 IU/mL is now generally used in the Bowman37 reported results on 997 pregnant women.
United Kingdom to indicate the need for invasive He made the following observations:
tests such as percutaneous umbilical blood sampling
(PUBS).1,2 1. A single ΔOD450 reading of 0.400 or higher at any
age of gestation is associated with the presence of
hydrops fetalis 65% of the time.
PREVIOUS HISTORY OF PREGNANT WOMAN 2. Occasionally, hydrops might be present at 28 weeks
of gestation when the ΔOD450 reading is 0.200–0.250.
If an expectant mother has antibodies (e.g., anti-D), it 3. ΔOD450 reading at the 80%–85% level in zone 2 can
is rare for HDFN to occur in the first pregnancy. Each
advance to a zone 3 reading and the presence of
successive D-positive baby is more likely to suffer
hydrops within 2 weeks.
with increasingly severe HDFN.2 If hydrops occurs
4. Rapid increases in ΔOD450 and severity of Rh
due to anti-D, there is a 90% chance that the next D- disease can occur. Thus, a reading in low zone 2 at
positive fetus will die in utero.2 23 weeks might be followed by a reading in zone 3
2 weeks later, with the presence of an hydropic
AMNIOTIC FLUID ANALYSIS fetus at the time of the second amniocentesis.
5. On rare occasions, initial ΔOD450 readings of
As early as 1950, Bevis35 used amniotic fluid to predict 0.200–0.250 at 22–24 weeks’ gestation might be
the severity of HDFN. Analysis of a bilirubin pigment associated with an Rh-negative fetus.
in amniotic fluid became popular after publication of
a method by Liley36 in 1961. Amniotic fluid, protected Queenan and colleagues38 pointed out that Liley
from the light that destroys bilirubin, was centrifuged charts started at 27 weeks’ gestation and that pro-
and filtered. Optical density (OD) measurements jected values before this time were inaccurate. He pre-
above the 350–700 nm range were plotted on semilog- sented a revised Liley chart (Fig. 13-1), which is now
arithmic paper using wavelength as the horizontal the one commonly used.
linear coordinate and OD as the vertical logarithmic In 1996, the American College of Gynecology
coordinate. The plotting readings were corrected, and (ACOG)39 recommended that amniocentesis or PUBS
the deviation from linearity at 450 nm (OD450) was should be considered if the antibody titer is 32 or

0.20 0.20
Intrauterine death risk
0.18 0.18

0.16 0.16
Rh positive
(affected)
0.14 0.14

0.12 0.12
ΔOD450 nm

0.10 Indeterminate 0.10

0.08 0.08

0.06 0.06
Rh negative
0.04 (unaffected) 0.04

0.02 0.02

0.00 0.00
14 16 18 20 22 24 26 28 30 32 34 36 38 40
Weeks gestation

FIGURE 13-1. Amniotic Fluid ΔOD 450 management zones. (From Queenan JT, Tomai TP, Ural SH, et al: Deviation in amniotic fluid optical density at a
wavelength of 450 nm in Rh-immunized pregnancies from 14 to 40 weeks gestation: A proposal for clinical management. Am J Obstet Gynecol
1993;168:1370–1376.)
524 Immune Hemolytic Anemias

greater. If a patient has had a prior affected pregnancy OTHER METHODS FOR ASSESSING AND
(neonatal exchange transfusion, early delivery for PREDICTING THE SEVERITY OF HDFN
alloimmunization, or intrauterine transfusion), anti-
body titers are not necessary because amniocentesis or Ultrasound. The pathophysiology of HDFN pre-
percutaneous umbilical cord blood sampling will be dicts that fetal anemia will trigger exaggerated ery-
required. The timing of the initial procedure is thropoiesis, resulting in hypertrophy of the placenta
determined by past clinical history. It is usually per- and fetal liver or spleen. The placenta will become
formed at least 4 to 8 weeks earlier than the prior ges- edematous and the liver parenchyma congested. The
tational age at which significant morbidity occurred, obstructive hepatomegaly and hypoproteinemia,
with some clinicians beginning at 20 weeks or even together with high-output congestive heart failure,
earlier. eventually result in fetal abdominal ascites followed
Readings in zone III (the uppermost zone) suggest by scalp and limb edema, hydrothorax, and pericar-
severe hemolytic disease with a high probability of fetal dial effusion. In its terminal stage, the hydropic fetus
death within 7 to 10 days. Readings in zone I (the could demonstrate cardiomegaly. Some of these
lowest zone) are reassuring, although neonatal changes can be shown using ultrasound. Weiner and
exchange transfusion could be necessary occasionally. coworkers41 found that ultrasound was slightly more
There are no reliable data concerning the optimal fre- accurate than amniotic fluid analysis in predicting
quency for repeated sampling. In general, amniocente- moderate to severe HDFN. In their series of 57 gravi-
sis is repeated every 1 to 4 weeks if the ΔOD450 das (including three twin pregnancies), amniotic fluid
measurement is in zone II (middle zone) and every analysis correctly predicted an affected or healthy
3 to 4 weeks if it has dropped into zone I. The frequency neonate in 79% of the cases, with three false-positive
depends on where in zone II the value falls and the predictions. Real-time sonography accurately pre-
pattern established in previous procedures. Declining dicted the clinical course in 86% of the cases, with no
values are encouraging, although they do not preclude false-positive predictions. The authors found that the
mild hemolytic disease. Stable or rising ΔOD450 meas- most useful ultrasound findings were serial measure-
urements are causes of concern.39 ments of the fetal abdominal circumference and fetal
Patients with results in zone I or low zone II can be liver size. Others have not found ultrasound to be so
allowed to proceed to term, at which point labor valuable.42,43 Nicolaides and asociates43 correlated
should be induced. In most cases, patients in the ultrasound findings and the degree of fetal anemia as
middle of zone II can progress to 36–38 weeks’ gesta- assessed by fetal blood sampling. Ultrasound was
tion, at which time delivery should be accomplished, found to be unreliable in predicting fetal anemia in the
by induction of labor if possible. Depending on gesta- absence of hydrops.43 More recently, Doppler ultra-
tional age, patients in zone III should either be deliv- sonography, which measures fetal hemodynamics,
ered or should receive intrauterine fetal transfusion. has been applied.
Similar considerations can apply to a rising titer in Fetal Doppler Ultrasonography. Doppler ultra-
upper zone II. Delivery is often by cesarean birth sonography is a noninvasive method of studying fetal
because of an unfavorable cervix, but a trial of labor is hemodynamics.42 There have been many studies relat-
not contraindicated.39 ing Doppler blood velocity evaluations and fetal
In patients with a poor obstetric history or in whom anemia. The most useful results have resulted from
the initial titer is 16 or greater before 27 weeks, the studies of middle cerebral artery time-averaged mean
timing of the initial diagnostic procedure depends on blood velocity. Mari44 reported the results of a large
the severity of erythroblastosis in prior pregnancies, multicenter study involving 110 alloimmunized preg-
on ultrasound findings, and on the degree of titer ele- nancies. They found that moderate and severe fetal
vation. In more severely affected cases, many experts anemia could be detected by an increased peak veloc-
currently recommend that fetal blood sampling rather ity in the middle cerebral artery, with a sensitivity of
than amniocentesis be used to assess the fetus during 100% and a false-positive rate of 12%. Stefos and col-
the second trimester. This is particularly important if leages45 added confirmatory data. Nevertheless, in
the father is heterozygous for the offending antigen. 2002 Abdel-Fattah and Soothill42 reviewed applica-
At the time of the initial blood sampling, the fetal tions of ultrasound (including Doppler) to HDFN and
hematocrit and antigen status can be determined. If concluded that although the information from the
the fetus is anemic, transfusion is indicated at that fetal aorta blood velocity measurements resulted in an
time. If the fetus is not anemic and the relevant improvement of the prediction equation, the morpho-
antigen is found to be present on the fetal blood cells, logical measurements were of no benefit. They con-
the timing of future diagnostic procedures will cluded that careful attention to the woman’s obstetric
depend on history, serial ultrasound findings, and the history and serial antibody quantitation is equally
hematocrit at the time of the last sample. If the fetus is important to the use of Doppler ultrasonography.
found to be antigen negative, no further procedures Fetoscopy, Percutaneous Umbilical Blood Sampl-
are necessary.39 ing (Cordocentesis/Funipuncture), and Chorionic
Figure 13-2 shows a management scheme suggested Villus Biopsy. Fetoscopy is a technique in which the
by Moise.40 second-trimester fetus can be visualized directly and
Hemolytic Disease of the Fetus and Newborn 525

Early ultrasound to determine


Figure 13-2. Algorithm for gestational age of an Rh-negative woman whose husband was of the
overall clinical management of
rhesus; OD = optical density; probable R2r (cDE/cde) genotype. The woman had
the sensitized gestation. Rh = Determine paternal anti-D quantitated at 40 IU/mL at 10 weeks’ gestation;
MoM = multiples of the median. serotype and genotype
MCA = middle cerebral artery; fetal blood sampling was not available. The couple,
From: Moise J. Management of
rhesus alloimmunization.
Rh negative
with one living child, were desperate for a second
Obstet Gynecol 2002, page Rh positive
607. child. They requested repeated plasmapheresis, which
Obtain serial maternal No further studies was performed on 54 occasions until delivery fol-
antibody titers necessary
lowed premature rupture of the membranes at 35
Critical titer reached weeks’ gestation; the baby was Rh-negative and the
DAT was negative at birth. In 59 Rh-immunized preg-
Heterozygous paternal Homozygous paternal
nancies, Nicolaides and colleagues49 compared the
phenotype phenotype predictive values of the ΔOD450 of amniotic fluid and
the hemoglobin level of fetal blood obtained by fetal
Amniocentesis for fetal
genotype testing
blood sampling. Studies were performed at 18–25
weeks’ gestation. The authors found a significant
RhD-negative fetus RhD-positive fetus
linear correlation between the degree of fetal anemia
and ΔOD450 of amniotic fluid; however, the values
No further studies
necessary
Begin serial amniocenteses for
OD450 or serial MCA Dopplers
were widely scattered, limiting their predictive value.
There was no ΔOD450 level that clearly separated
OD450 value in upper severely from mildly affected babies. If the cutoff level
zone 2 or zone 3 of Liley
curve or intrauterine
was set low enough to include 90% of the severely
transfusion zone of
Queenan curve or MCA
affected babies (e.g., >0.2 nm), the group would
Doppler > 1.5 MOM's also include 54% of the mildly affected babies.
Alternatively, if the cutoff level was set high enough
Fetal blood sampling
(>0.5 nm), the false-negative rate was 77%. There was
an overall linear correlation between fetal hemoglobin
Fetal hematocrit <30% Fetal hematocrit >30%
and OD450. The normal range of fetal hemoglobin was
Assess gestational age
found not to change within the gestational range of
16–25 weeks. Although the predictive value of the
> 35 weeks < 35 weeks
ΔOD450 in the third trimester has been said to be
Delivery Intrauterine transfusion
94.5%, the data of Nicolaides and colleagues49 cast
serious doubt on applying the same logic to the
FIGURE 13-2. Flow chart showing a plan for managing the first
affected pregnancy. (From Moise KJ Jr: Changing trends in the
second trimester. Of the 31 most severely anemic
management of red blood cell alloimmunization in pregnancy. Arch fetuses (hemoglobin <6 g/dL), which included 14
Pathol Lab Med 1994;118:421–428.) fetuses with hydrops fetalis, only 10 (32%) would
have received intrauterine transfusions at the time of
amniocentesis had the “extrapolated” Liley criteria
fetal blood (or other tissues) can be sampled through an been used.49 Nicolaides and colleagues49 conclude by
endoscope introduced transabdominally into the am- suggesting that although fetal blood sampling could
niotic cavity.46 The technique is only reliably successful be associated with a marginally greater risk of fetal
at 16 weeks’ gestation and later.47,48 There is about a 5% morbidity than amniocentesis, the only accurate
mortality rate due to fetoscopy, compared with a method to assess the severity of fetal anemia in the
1%–2% mortality rate after midtrimester amniocente- second trimester is to measure the hemoglobin level of
sis.47,48 The fetal blood can be tested for blood type, fetal blood. In 1983, Daffos and coworkers50 reported
DAT, hemoglobin, and hematocrit. MacKenzie and a new technique for obtaining fetal blood that
coworkers47,48 suggested that the technique would be involved umbilical cord sampling with an ultrasoni-
of benefit in cases in which the father of the baby is cally guided 20-gauge needle. They later reported a
known to be heterozygous for the offending blood series of 606 fetal blood samplings in 562 pregnancies
group antigen in the following situations: from 17 to 38 weeks’ gestation.51 Their reported com-
plications were premature delivery (5%), growth
1. Patients with a history of previous babies having retardation (8%), in utero death (1.1%), and sponta-
severe HDFN neous abortion (0.8%). This PUBS is now more
2. Patients with high-titer anti-D (e.g., in excess of popular than blood sampling using fetoscopy.52
20 IU/mL) during the first 20 weeks of pregnancy Fetoscopy and PUBS have been used to obtain
blood for studying the presence and development of
The authors felt that knowledge that the fetus was blood group antigens on fetal RBCs. These have
blood group incompatible would save considerable included ABO and Rh and 30 other blood group anti-
time, effort, and expense by obviating the need for gens.53,54 Habibi and colleagues54 confirmed that I,
further diagnostic tests and exacting methods of treat- Lea, and Leb are not expressed on fetal RBCs and that
ment. MacKenzie and colleagues47 gives an example the expression of A, B, H, P1, P, Lua, and Lub antigen
526 Immune Hemolytic Anemias

is decreased compared with adults. Many antigens, In Vitro Predictive Tests Utilizing Functional
including D, C, c, E, e, K, k, Kpa, Kpb, Fya, Fyb, Jka, Jkb, Cellular Assays. In vitro functional cellular assays,
M, N, S, s, Xga, Vea, and Gea, appeared to be fully based on the in vivo mechanisms of RBC immune
developed by the second half of gestation. The P destruction, have been developed and applied to pre-
antigen was found to be absent from 12% and weakly dicting the clinical significance of alloantibodies
expressed on 10% of the fetal samples, which were involved in HDFN and hemolytic transfusion reac-
constantly P1 negative. Some investigators have used tions.62-76 These assays are based on sensitizing RBCs
a technique known as chorionic villus sampling with the putative antibody and adding these sensi-
(CVS) to obtain fetal blood samples during the first tized RBCs to monocytes in vitro. The interactions
trimester of pregnancy. These investigators have with the monocytes are measured by recording RBC
studied ABO, Rh, and K antigens on fetal RBCs from adherence/phagocytosis by a monocyte monolayer
CVS between 8 and 12 weeks’ gestation.55-57 They assay (MMA), antibody-dependent cellular cytotoxic-
suggest that such studies could be useful in predict- ity (ADCC) using 51Cr-labeled RBCs, or a chemilumi-
ing the clinical course of a fetus of an immunized nescence test (CL) using luminol. The MMA and CL
mother, but on the other hand, there is a total have been found to be equivalent to performing a
pregnancy loss rate of 0.6–0.8% higher than with 1-hour 51Cr-RBC survival study when evaluating the
amniocentesis.58 possibility of transfusing incompatible blood.
Prenatal Determination of Fetal Blood Group. For Application of these assays to predicting severity of
many years, the babies’ blood group was determined HDFN has been more controversial (see the discus-
as an educated guess. The process involved serologi- sion later in this chapter).
cal testing of the mother and father and evaluating In 1995, some international experts reviewed labo-
the heterozygosity vs. homozygosity of the father by ratory procedures that were found useful in predict-
relating to published data of the probability of phe- ing the severity of HDFN.65 They were asked: “Is a
notype results representing a certain genotype. The laboratory procedure used in your country/center to
father’s result was reported as a “probable genotype” predict the severity of HDFN?” If so:
(Table 13-7). If the father was thought to be homozy-
gous, the baby was assumed to possess the putative 1. Which assay is used?
antigen. If the father was heterozygous, there was a 2. Why was this assay selected?
50% chance that the baby was antigen positive. With 3. At which time during pregnancy is the assay
the advent of PUBS, it was possible to obtain RBCs performed?
from the fetus to type the fetus using routine sero- 4. How are the results interpreted, and what are the
logy. It is now possible to determine the Rh(D,c,E), K, consequences for the management of the case?
Fya, and Jka status of the fetus accurately, applying
molecular techniques to the fetal RBCs, amniocytes, If not:
or even the maternal plasma.59–61 Molecular tech-
niques can also be used to determine the father’s true 1. How are known cases of maternal alloimmuniza-
genotype. Although this is the modern approach, tion managed; that is, how is the status of the fetus
readers should be warned that results should be controlled?”
relied on only if they come from laboratories with a
trusted knowledge base in the complexity of the Rh The main conclusion from the answers received was
system at the DNA level. that in assessing the clinical significance of maternal

TABLE 13-7. ESTIMATES OF THE LIKELIHOOD OF HOMOZYGOSITY FOR D IN CAUCASIANS

Phenotype Anti-D Anti-C Anti-c Anti-E Anti-e Probable Genotypes* Likelihood of Homozygosity for D

R1r + + + – + DCe/dce R1r 4.8%


DCe/Dce R1R0
R1R1 + + – – + DCe/DCe R1R1 97.6%
DCe/dCe R1r’
R2r + – + + + DcE/dce R2r 4.8%
DcE/Dce R2R0
R1R2 + + + + + DCe/DcE R1R2 96.7%
DCe/dcE R1R”
dCe/DcE r’R2
R2R2 + – + + – DcE/DcE R2R2 96.6%
DcE/dcE R2r”

*Rare alternative genotypes have not been shown.


Hemolytic Disease of the Fetus and Newborn 527

anti-D, functional cellular assays are not used rou- the previous pregnancy, the ADCC test is done for the
tinely except in the Netherlands. In all other countries, first time at the 20th week. If the results in a previous
determination of the IAT titer (and in most cases, the pregnancy were predictive of a severe HDFN, the
determination of the concentration of anti-D using an assay is performed between weeks 12 and 14. Based
AutoAnalyzer) are the routine procedures used. If the on the analysis of a large number of cases (n = 482),
titer is above a critical level (mostly >16–32) or the the results of the tests were interpreted as follows:
anti-D concentration is greater than 4–5 IU, special
attention is given to the status of the fetus, and if the • ADCC lower than 30%: No hemolysis expected; no
concentration is greater than 10–15 IU, the case is amniotic fluid analysis; after week 32, repeat
usually referred to a center specializing in the man- ADCC test every 2 weeks; after week 36, repeat
agement of HDFN. Several answers stressed that an ADCC weekly; before week 32, repeat ADCC test
increase in anti-D titer or concentration during preg- every 3 to 4 weeks.
nancy is of greater significance than the actual titer or • ADCC 30%–50%: No hemolysis or moderate
concentration. In the Netherlands, the ADCC test with hemolysis expected; fetus control by ultrasonogra-
monocytes as effector cells is routinely applied in the phy; amniotic fluid analysis, but frequency to
Central Laboratory of the Netherlands Red Cross depend on the results of the first analysis; after
Blood Transfusion Service (now known as the week 32, repeat every 2 weeks; after week 36,
Sanquine Blood Supply Foundation) for the whole repeat ADCC weekly; before week 32, repeat every
country.65,68 The results of the test determine whether 3 weeks.
cordocentesis is indicated and whether the case • ADCC greater than 50%: Severe hemolysis
should be referred to a specialized center.65 In some expected; patient referred to a center specialized in
other countries, a functional assay is used in special the management of HDFN.
cases (e.g., if in a previous pregnancy the severity of
the HDFN was much worse than expected from the Oepkes and colleagues68 compared the ADCC assay
anti-D titer or concentration, or when there is a rapid with antibody titers and found that the ADCC assay
increase in anti-D).68 consistently showed a higher predictive value for fetal
It now seems generally accepted, based on the disease than did the AGT titer.
results of several comparative studies, that the ADCC Monocyte Monolayer Assay. Nance and her Los
test and the CL test with monocytes as effector cells Angeles colleagues69,70 compared the efficiency of the
offer the best predictive value for severity of HDFN, MMA with that of titers and amniotic fluid OD450, in
when using functional cellular assays. predicting the severity of HDFN. Maternal sera were
Antibody-Dependent Cellular Cytotoxicity. taken at birth or at the time of amniocentesis, from
Urbaniak and coworkers67 used a lymphocyte- women with antibodies of potential clinical signific-
driven ADCC assay to predict the severity of HDFN. ance in their sera. Cord sera, DAT-positive cord RBCs,
They sensitized homologous enzyme-treated group and eluates from these RBCs were also tested.70 The
O Rh-positive RBCs with maternal antibody and sera and eluates were used to sensitize allogeneic
used these as target cells in the ADCC assay, utilizing group O RBCs having the reactive antigen (e.g., D).
normal donor lymphocytes as effector cells. They The sensitized RBCs were added to a monocyte mono-
described three cases in which amniotic fluid OD450 layer prepared from normal donor. Thirty-one cases
(Liley low zone in cases one and two and mid-zone with anti-D, 12 cases with other Rh antibodies (six
in case three) predicted a favorable outcome anti-c, two anti-C, one anti-E, two anti-c+E, and one
although maternal anti-D levels predicted a poor anti-c+s), and seven cases with non-Rh antibodies
outcome (26, 20, and 50 IU/mL, respectively); the (two anti-K, one anti-S, one anti-Jka, one anti-Jkb, one
ADCC assay results were positive in cases 1 and 3 anti-Wra, and one anti-Dib) were studied.70 The babies
and negative for case 2. Cases 1 and 3 needed were divided into five groups. Group 1 (n = 4) were
exchange transfusion at birth, whereas case 2 asymptomatic; group 2 (n = 21) had bilirubin levels
required only “top-up” transfusion. In five cases in greater than 5 mg% at 48 hours but required no
which antibody levels were high and OD450 pre- treatment; group 3 (n = 6) required phototherapy for
dicted the outcome correctly, the ADCC assay was jaundice; group 4 (n = 6) required IUT or neonatal
also positive. In two cases in which, despite high transfusions; group 5 (n = 4) were fatal cases. The
anti-D levels, the babies required no treatment, MMA results using maternal sera were better than
extremely low ADCC assay activity was observed. In the others at predicting whether babies would need
one case, the baby was Rh negative and the OD450 treatment (groups 3, 4, and 5). As might be expected,
was high mid-zone, indicating an affected baby; the the maternal sera gave slightly more false-positive
ADCC assay also gave a false-positive result. results (e.g., in one case of an Rh-negative infant
In the Netherlands, an ADCC assay using mono- whose mother’s serum contained high-titer anti-D).
cytes rather than lymphocytes is performed in a The antibody titer was better than some other
central laboratory.65,68 When anti-D is detected for the studies have suggested, with an efficiency of
first time, the ADCC assay is first done during the 72%–80%; the efficiency of the MMA was 83%, but
32nd week of pregnancy. If anti-D was found in the predictive value of a negative test was 94% (16 of
528 Immune Hemolytic Anemias

17 negative MMAs were associated with a benign clin- nized pregnancies (51 anti-D, eight –D,C, six –c, six
ical course). As the MMA showed a reasonable corre- –E, 2 – cE, two –K, and one each anti-D,E, -D,C,E,
lation with the clinical status of the baby at birth, a -D,C,s, -C, -Cw, -e, -Jkb, -Wra, -Dib, -U, -Ge) who deli-
further study was performed to see how the MMA vered antigen-positive infants. Seventy-nine percent
would compare with amniotic fluid analysis during of the maternal sera contained IgG1 antibodies (31%
the pregnancy in predicting severity of HDFN.70 IgG1 only); 16% contained IgG2 (1% IgG2 only); 62%
Parallel studies of amniotic fluid OD450 and MMAs contained IgG3 (10% IgG3 only); and 12% contained
were performed on 29 sera from 16 pregnant women IgG4 (0% contained IgG4 only).
with Rh antibodies (nine anti-D, three anti-C+D, two The correlation of IgG subclass and the severity of
anti-E, one anti-E+D, one anti-c). The predictive value HDFN is controversial. Schanfield81 presented a small
(PV) of positive OD450 result (Liley zone mid-2 to -3) amount of data that suggested that neonates born
was 100%, but the PV of a negative result was only with RBCs sensitized with IgG1 Rh antibodies had
60%; the efficiency of the OD450 was 75%. The PV of a lower mean cord hemoglobin and higher cord biliru-
positive MMA was 91% and the PV of a negative bin levels, with a lower postnatal bilirubin rise, than
MMA was 100%, giving an efficiency of 94%. These neonates with IgG3-sensitized RBCs. He suggested
results suggested that the MMA would be a very that this finding related to the preferential placental
useful test for predicting the severity of HDFN. It was transfer of IgG1, which allowed for a longer period of
noninvasive and thus did not carry any risks to the in utero IgG1 sensitization. In contrast, neonates with
mother or baby. Its only disadvantage seemed to be an IgG3 sensitization tended to have higher mean cord
occasional false-positive result (e.g., when the mother hemoglobin and lower cord bilirubin levels, but a
is carrying an Rh-negative baby and has Rh antibodies higher postnatal rise in bilirubin. This was thought to
in her serum). The assay seemed to be a good adjunct be due to the shorter in utero exposure to IgG3 but the
to other noninvasive approaches such as ultrasound. relative higher efficiency of IgG3 in causing postnatal
Larson and associates74 agreed with the findings of RBC destruction. Schanfield81 also suggested that
Nance and colleagues70 that the MMA yielded a high within the IgG1 subclass, G1m(a) antibodies were
PV (a negative PV of 100% and a positive PV of 87.5%). significantly less effective than G1m(f). Mothers of
In contrast, Moise and coworkers75 found there was no moderately and severely affected infants showed a
difference in MMA results between anemic and striking absence of G1m(a+) IgG1Rh antibodies; the
nonanemic fetuses. In two later studies by the Los majority of antibodies in this group were G1m(f),
Angeles group, the MMA was found to only have an G3m(g), or mixtures of IgG1 and IgG3. It was sug-
efficiency of and 66% and 57%, respectively.62,71 The gested that the differences in transport of subclass-
two latter studies included antibodies other than anti- specific antibody across the placenta and their
D and babies who lacked antigens reactive with the hemolytic potential may account for some of the dis-
maternal antibody; in addition, there were less severely crepancies noted between maternal antibody titers
affected babies than in the earlier studies.62,70 Similar and severity of HDFN. For example, a mother with an
findings were reported by Zupanska and collea- Rh antibody (titer 2048) that is totally IgG3 might
gues,72,73 who at first reported the MMA to be efficient deliver an infant that is not severely affected. The
in predicting the severity of HDFN but later found that child would probably need therapy postnatally,
the assay failed when moderate disease was present. however. In contrast, an IgG1, G1m(f) antibody of
Chemiluminescence Test. The CL has been com- similar titer could cause severe HDFN in utero,
pared with ADCC, MMA, and antibody quantitation leading to a stillborn fetus. Rh IgG1 antibody of lower
in predicting the severity of HDFN.76 The CL titer (e.g., 64–128) might be associated with a mildly
appeared to be equivalent to the ADCC assay and affected infant at birth, requiring no further treatment.
more efficient than MMA and antibody quantitation. An IgG3 antibody of the same titer, however, might be
IgG Subclass. Most IgG Rh antibodies formed associated with an infant who is mildly affected at
during pregnancy are IgG1 and IgG3. Frankowska birth but subsequently becomes severely affected. In
and Gorska77 found that 87.6% of sera from 65 preg- summary, Schanfield81 suggested that IgG1 antibodies
nant women contained IgG1 Rh antibodies, 23% con- caused greater destruction in utero followed by a
tained IgG2 antibodies, 56.9% contained IgG3 more benign clinical course; in contrast, IgG3 antibod-
antibodies, and 7.7% contained IgG4 antibodies. Most ies appeared to cause less destruction in utero but
commonly, the sera contained IgG1 alone (33.9%) or were associated with a much more severe clinical
IgG1 + IgG3 (32.3%); no sera contained IgG2 and/or course postnatally. Unfortunately, these data have
IgG4 without IgG1 or IgG3. Ouwehand and associ- never been extended, and no one has confirmed the
ates78 studied 52 isoimmunized D-negative women; hypothesis thus far.
35% of them produced only IgG1 antibodies, 29% pro- Frankowska and Gorska77 could show no correla-
duced only IgG3 anti-D, and 39% produced both IgG1 tion between IgG subclass and severity of HDFN.
and IgG3. Parinaud and colleagues79 found that IgG1 Parinaud and coworkers79 found that IgG1 Rh anti-
and IgG3 Rh antibodies were equally represented bodies were associated with higher neonatal biliru-
(present in 59% of sera) in 103 isoimmunized women. binemia than IgG3 antibodies; within the IgG1 group,
Nance and coworkers80 reported on 86 alloimmu- the G1m(4) allotype was associated with the most
Hemolytic Disease of the Fetus and Newborn 529

severe HDFN. Severe HDFN appeared to be less families in which Rh HDFN had occurred and found
common when IgG1 and IgG3 were present together; that the immunizing pregnancy was always ABO
it was suggested that IgG3 might exert a protective compatible. These and other observations stimulated
effect. Cregut and associates82 found that IgG3 Rh a group of workers in Liverpool, England, to suggest
antibodies were associated with less severe HDFN. In that perhaps the phenomenon could be mimicked,
contrast, Taslimi and colleagues83 found that HDFN when the mother and fetus were ABO compatible, by
occurred only when IgG3 Rh antibodies were present. giving an antibody to destroy the Rh-positive fetal
Nance and coworkers80 found that when IgG1 anti- RBCs in the maternal circulation.93 These workers
bodies were present without IgG3 antibodies, 56% of went on to demonstrate, using the Kleihauer-Betke
the babies needed treatment (8% phototherapy, 26% fetal cell staining technique, that the higher the fetal
transfusion) and 2% had fatal HDFN. When IgG3 was cell score at delivery, the more likely the women were
present without IgG1, 47% needed treatment (20% to become immunized.94 They also tried to give anti-D
phototherapy, 27% transfusion). When IgG1 and IgG3 to Rh-negative volunteers who had been injected with
were both present, 90% required treatment (21% pho- 51Cr-labeled Rh-positive RBCs.95 Unfortunately, this
totherapy, 63% transfusion), and 5% had fatal HDFN. experiment was a failure, as plasma containing IgM
Several other reports have agreed that IgG1 alone and anti-D was used. Fortunately, they were stimulated to
IgG1 with IgG3 have a higher association with sever- repeat the experiment, with plasma containing IgG
ity of HDFN than IgG3 alone.84-89 anti-D,96 when they heard of work by Stern and
Lambin and associates89 found that the median per- coworkers in a Chicago group.97,98 Stern and cowork-
centages of IgG1 and IgG3 anti-D in maternal sera ers had shown that Rh-negative volunteers failed to
were 90% and 10%, respectively, whereas on infants’ make anti-D when they were injected with Rh-posi-
RBCs they were 97% and 3%, respectively. The differ- tive RBCs coated in vitro with IgG anti-D. In 1961 and
ences between maternal and infantile percentages 1963, the Liverpool group published the impressive
were significant (P < 0.001). IgG1 and IgG3 anti-D results of their experiments.95,96,99 Only three of 21 Rh-
bound to infants’ RBCs increased concomitantly with negative volunteers made anti-D after receiving Rh-
the concentration of IgG1 and IgG3 anti-D in the positive RBCs if they also received plasma containing
maternal sera. The severity of HDFN correlated posi- IgG anti-D; this was in contrast to the 21 controls who
tively with the concentration of IgG1 anti-D in mater- did not receive anti-D, in which group 11 were sensi-
nal sera, but negatively with the amount of IgG3 tized to produce anti-D. Independently, another
anti-D bound to infants’ RBCs. In addition, the exist- group in the United States was working on the same
ence of a high proportion of IgG3 anti-D in maternal problem. This group had been stimulated by the early
serum was associated with a delayed risk of fetal observations of Smith,100 who, in 1909, had shown
anemia. Thus, the proportion of IgG3 anti-D relative that antigen-antibody complexes of diptheria were
to the total IgG anti-D on infants’ RBCs is only one not immunogenic when the complexes were prepared
third the proportion present in maternal sera. The in antibody excess. Freda and Gorman101 thought that
study of the correlations between the amount of IgG1 if an excess of passive antibody (e.g., anti-D) was
and IgG3 anti-D and the severity of HDFN suggests given to an individual, then subsequent injections of
that IgG1 anti-D was found to be more important than antigen (e.g., O+ red cells) would fail to cause sensiti-
IgG3 anti-D in the pathogenesis of fetal anemia. zation, the antibody acting as part of the humoral
Although there is a statistical correlation between homeostatic mechanism and exerting a negative
severity and IgG subclass, as illustrated in the forego- “feedback” on antibody synthesis. Freda and
ing discussion, there is so much variation among indi- colleagues102 carried out a trial in which they injected
vidual cases (e.g., strong IgG RBC sensitization Rh-negative prisoners in Sing Sing prison with an IgG
associated with no hemolysis in one baby and severe fraction containing anti-D after the prisoners had
hemolysis in another baby), that subclassing has no received Rh-positive RBCs. This approach was emi-
practical value in predicting the severity of HDFN nently successful. On day 1 of the trial, 27 Rh-negative
when testing maternal sera before birth. men received intravenous injections of Rh-positive
RBCs (10 mL). Then, 72 hours later, 14 men received
intramuscular injections of a sterile hyperimmune
RH IMMUNE PROPHYLAXIS anti-Rh gamma globulin preparation (Rh Ig). At
6 months after the procedure, none of the 14 men who
In 1943, Levine90 pointed out that in matings in which received Rh Ig had produced anti-D, but 6 of the 13
an Rh-negative mother carried an ABO-incompatible controls were immunized. After the success of their
fetus, there were significantly fewer cases of Rh experiments, both the British and American groups
HDFN. Later, Race and Sanger91 speculated that the went ahead with clinical trials. A combined study
mechanism of this protection might be that the Rh- from British centers and Johns Hopkins Hospital in
positive ABO-incompatible cells of the fetus were Baltimore103 in 1966 and a study by Pollack and asso-
rapidly destroyed on entering the mother’s circula- ciates104 in 1968, clearly showed the efficiency of Rh Ig
tion and thus were incapable of initiating Rh sensiti- in preventing Rh sensitization, with virtually 100%
zation. In 1956, Nevanlinna and Vainio92 examined prevention of sensitization being achieved in women
530 Immune Hemolytic Anemias

who received Rh Ig. One of the main reasons the clin- massive FMH will lead to a failure of Rh immuniza-
ical trials in the United States were so successful with tion prevention in less than 0.1% of women at risk.
no adverse reactions was the development of a suit- As early as 1967, Zipursky and Israels107 noted that
able Rh Ig by the Ortho Research Foundation in the main cause of failure in Rh immunoprophylaxis
Raritan, New Jersey. The original preparation was a was sensitization during pregnancy and that the only
gamma globulin fraction of pooled sera from a small way to prevent this was antenatal prophylaxis. In a
number of Rh-negative donors who had been hyper- series reported by Bowman and colleagues,108 Rh
immunized with Rh-positive RBCs. The 16.5% gamma immunoprophylaxis was predicted to fail in 1.6% of the
globulin solution was pure IgG (free from IgM) and Rh-negative women if Rh Ig was given only after deli-
was sterilized and packaged in 5-mL vials, suitable for very. After administration of Rh Ig at 28 and 34 weeks
intramuscular injections. This product led to the of pregnancy, in addition to a further administration
highly standardized successful product RhoGAM, within 72 hours of delivery, the failure rate was only
which was licensed by the FDA in 1968.105 The stan- 0.07%. At a conference in 1978, other workers, includ-
dard dose for clinical use in the United States is ing those from Europe, the United States, and Canada,
300 μg, but smaller doses seem to be equally effective showed that of 1078 prior gravidas and multigravidas
and are used in other countries such as England and treated antenatally, only three (0.37%) showed evidence
Australia (100 μg and 125 μg, respectively).2 of immunization, compared with 39 of 2550 (1.53%) of
women treated only postnatally.109 Although the data
PROBLEMS IN COMPLETELY ERADICATING just presented sounds impressive, antenatal prophy-
Rh HDFN laxis remains controversial, especially in some coun-
tries.110-117 Cost-benefit issues are the primary concerns.
Although Rh Ig was a highly successful approach to Some workers in the United Kingdom and the United
eradicating Rh HDFN, it became obvious that failures States have argued that it is not worth the national
were occurring. Some of these were due to obvious expense of giving all Rh-negative women Rh Ig ante-
causes, in that some Rh-negative women were not natally in addition to postnatally; other workers in the
receiving Rh Ig, and this will probably always occur. same countries have argued for antenatal Rh prophy-
Among the group receiving Rh Ig, however, there laxis.114-117 Mollison117 makes the point that it is impor-
was still a small number of failures. After about a tant to make the distinction between the prevention of
decade of Rh Ig usage, the failure rate (i.e., an Rh- Rh immunization and the prevention of Rh HDFN. For
negative mother making anti-D in a pregnancy instance, if a woman is primarily immunized only by
subsequent to delivering an Rh-positive baby and her second pregnancy and if she has no further preg-
receiving Rh Ig), was approximately 1%. The nancies, then virtually no harm has been done.
main causes of these failures were shown to be the Mollison117 suggests that in these days when most
following: women are content to have just two children, one can
make a good argument for giving antenatal prophy-
• Rh-negative women who had aborted106 laxis during the first pregnancy only. If antenatal Rh Ig
• Rh-negative women who had amniocentesis106 is withheld during the following pregnancy despite
• Rh-negative women with a massive fetal maternal there being a 1% chance of Rh immunization, routine
hemorrhage (FMH)106 testing for antibody, premature delivery, and exchange
• Rh-negative women immunized during pregnancy93 transfusion in the very few cases in which the infant is
severely affected, should result in negligible mortality.
These associations led to recommendations that a Mollison117 makes a final point that antenatal prophy-
smaller dose of Rh Ig (e.g., 50 g) be given to all Rh- laxis involves waste, not only because it is superfluous
negative women who abort or undergo amniocente- in most cases (in which postnatal treatment alone
sis. Because the smaller dose effectively suppresses would suffice), but also because in about 40% of cases
only the response to 2.5 mL of Rh-positive red cells, it is given to women who are carrying Rh-negative
some workers prefer giving a full dose after amnio- infants.
centesis, but other workers have suggested that Rh Ig
is not necessary after early spontaneous abortion or
amniocentesis. The protective effect of Rh Ig is dose CLINICAL FEATURES OF HDFN
dependent. To be able to prevent Rh immunization
completely, for every 1 mL of Rh-positive blood The following features will be those associated with
(0.5 mL of RBCs), an Rh-negative woman would need HDFN due to Rh antibodies. HDFN due to ABO anti-
to receive 10 μg of Rh Ig. Thus, the 300 μg dose rec- bodies will be dealt with separately later in this
ommended in the United States should prevent chapter. Features of most other IgG alloantibodies,
immunization when the FMH is less than 30 mL of with the possible exception of anti-K (see earlier), will
blood (15 mL of RBCs). An FMH greater than 30 mL be similar to those of Rh.
produces Rh immunization in about 35% of cases who HDFN is a result of the fetal RBCs becoming
receive only 300 g of Rh Ig; such cases occur in about sensitized with IgG antibody. Complement activation
0.2% of pregnancies. Thus, neglecting to screen for a rarely occurs and is not thought to be involved in
Hemolytic Disease of the Fetus and Newborn 531

the pathogenesis of the disease (even in ABO HDFN at 10–20 days after birth; the anemia is not
HDFN).2,118,119 The IgG RBCs are removed extravascu- accompanied by hepatosplenomegaly or jaundice. The
larly by the mononuclear phagocytic system (MPS); anemia seems to be an accentuation of the normal
intravascular complement-mediated lysis never occurs decrease in Hb/Hct that occurs normally at this time.
(even in ABO HDFN). The fetal MPS is probably This late anemia can occur in babies who have
capable of destroying sensitized RBCs before the fourth responded to exchange transfusion at birth or in babies
month of pregnancy, as intrauterine death from HDFN who required little or no treatment at birth. The late
has been recorded before the 20th week, although it is anemia is usually only moderately severe and rarely
uncommon before the 24th week. HDFN varies greatly needs treatment with transfusion, but occasionally the
in severity. It can be associated with a severe hemolytic anemia can be profound, requiring transfusion.2 The
process leading to intrauterine death. The fetus could cause(s) of the late anemia is uncertain and could have
die in utero at any time from the 18th week of gestation multiple origins (e.g., increased RBC destruction due to
onward. The incidence of stillbirth associated with Rh- alloantibodies and/or infection, decreased red cell pro-
HDFN varies according to the obstetric history of the duction due to alloantibodies (e.g., anti-K), erythropoi-
woman. In first affected infants, the stillbirth rate has etin deficiency, lack of response to erythropoietin,
been reported as 6%, but in second affected and later ineffective erythropoesis, and/or infection).
infants the rate can be as high as 29%. If a woman has Table 13-8 lists some of the clinical problems that
had only a mildly affected infant, her chance of a sub- can be associated with HDFN.
sequent stillbirth is probably as low as 2%, but if she
has had one previous stillbirth, the risk of subsequent ASSESSMENT OF SEVERITY OF
stillbirth is reported to be as high as 70%. In the most
severe cases, there is gross enlargement of the liver, DISEASE IN THE NEWBORN INFANT
spleen, and heart, and sometimes there are also ascites
and generalized edema giving rise to the condition A positive DAT cannot be equated with HDFN. Only
known as hydrops fetalis. About 20% of HDFN cases about 60% of infants with positive DATs due to Rh(D)
present with fetal hydrops. When born alive, infants sensitization require any form of treatment.2 There is
with severe HDFN could have cord hemoglobin (Hb) no direct correlation with the strength of the positive
concentrations of 3.5–8 g/dL; the number of nucleated DAT and the severity of HDFN, although the correla-
red cells could be as high as 200 × 109/L, and the retic- tion is similar to that discussed in Chapter 4 for AIHA:
ulocyte count could be as high as 60%.2 In such infants, Most cases of severe HDFN are associated with
jaundice can develop rapidly. Unless the infant is strongly positive DATs, and most (but not all) babies
treated promptly, kernicterus may develop. with weakly positive DATs have mild or no HDFN.
Kernicterus is a syndrome characterized by signs of Nevertheless, as with AIHA, there are many excep-
damage to the brain. About 70% of infants developing tions to these findings. Hughes-Jones and cowork-
kernicterus die between the second and fifth days of ers121 measured the amount of red cell-bound IgG Rh
life.2 At autopsy, these infants are found to have yellow antibody on cord RBCs and found 0.4–18 μg/mL. The
staining of the basal ganglia of the brain. Those that amount on the RBCs was not highly correlated (corre-
survive have permanent cerebral damage; some infants lation coefficient of 0.6) with the infants’ cord Hb or
show deafness as the only sign. There is a close rela- bilirubin concentrations. All infants with more than 8
tionship between the peak serum bilirubin concentra- μg/mL of antibody required treatment, but even at a
tion and the development of kernicterus. In mature level of 2 μg/mL, six of 14 infants required treatment.
infants, kernicterus rarely develops in association with Garratty and Nance122 used flow cytometry to
bilirubin concentrations of less than 18 mg/dL, examine the amount of IgG Rh antibody on DAT-
although in premature infants there is some evidence of positive newborns. Although babies with evidence for
kernicterus at lower bilirubin levels. More moderate
forms of the disease may present with less anemia and
bilirubinemia that respond to transfusions, and these TABLE 13-8. CLINICAL PROBLEMS THAT COULD
might not necessarily be exchange transfusions.2 BE ASSOCIATED WITH SEVERE HDFN
About 25% of HDFN cases are of moderate severity.
Infants with a mild form of HDFN (about 45%–50% of Intrauterine fetal demise
all HDFN cases) might present with little or no jaun- Anemia
dice at birth. Mild jaundice and anemia (e.g., slightly Thrombocytopenia
more rapid than the normal fall in Hb, remembering Kernicterus
that it is normal for the Hb to fall during the first few Coagulation defects (e.g., DIC)
Low serum albumin
weeks after birth) might develop after birth, and the Hypoglycemia
baby might need minimal treatment (e.g., photothe- Hypocalcemia
rapy) or no treatment.2 Pleural effusion, ascites, hydrops
Sometimes a late anemia is associated with Respiratory distress syndrome
Birth asphyxia
HDFN.2,120 This entity can present as the sudden Preterm delivery
appearance of anemia in an infant who was born with
532 Immune Hemolytic Anemias

HDFN had higher levels of RBC bound IgG, there was AMELIORATION AND TREATMENT
considerable overlap between the clinically affected OF HDFN
and nonaffected groups. No clear quantitative
hemolytic threshold was obvious. Nance and associ-
ates69,70 also correlated the results of an MMA (using There are many problems to deal with in attempting
the cord RBCs) with severity of HDFN. Once again, to salvage a child suffering with HDFN. Table 13-8
the MMA correlated quite well, but the correlation shows some of these problems.
was not perfect.
Mollison and colleagues2 reported that the cord Transfusion
blood hemoglobin concentration was the best single
criterion of severity; if the logarithm of the hemoglobin In 1950, Allen and associates124 demonstrated the value
concentration was plotted against the probit of the sur- of neonatal exchange transfusion in overcoming
vival rate, a linear relationship was observed. The cord anemia and preventing kernicterus. In 1954, the same
blood bilirubin concentration was of less value on its group showed the benefits of preterm delivery; the risk
own, but when taken in conjunction with the cord Hb of fetal death due to HDFN was reduced to 25% (from
concentration, it is of great value, especially in assess- 50%) of the immunized pregnancies.125 During the last
ing dangerously high bilirubin levels. Mollison and col- decade, there have been tremendous advances in the
leagues2 point out that the normal range (mean + 2 SD) care of preterm babies. Most major centers can now
of hemoglobin values in cord blood was approximately claim more than 80% survival rate at 28 weeks’ ges-
13.6–19.6 g/dL, whereas the range on venous blood tation. In 1961, Liley126 described a technique for
drawn in the first 24 hours of life is 14.5–22.5 g/dL. intrauterine transfusion (IUT). He transfused fresh Rh-
Before Rh prophylaxis, almost 50% of affected infants negative blood into the fetal peritoneal cavity,
had cord blood Hb concentrations of 14.5 g/dL or correcting the anemia and allowing the fetus to survive
greater, and 30% had values between 3.4 g/dL and until it was mature enough for preterm delivery.
10.4 g/dL. Most infants with cord Hb concentrations Experiences with this technique have varied. Robertson
within the normal range do not require exchange trans- and colleagues127 and Palmer and Gordon128 felt that
fusion.2 A level of 4 mg/dL or more of cord blood the value of the technique was questionable, Robertson
bilirubin (normal range, 0.7–3 mg/dL) or a rapidly and colleagues127 had a fetal mortality rate of 20% due
rising bilirubin is suggested as an indication for to trauma, and in a further 40% of the stillbirths, death
exchange transfusion.2 If the cord total bilirubin is less occurred within 48 hours of the IUT. Other centers have
than 4–5 mg/dL and rising only slowly, phototherapy reported more success with IUT.11,13,37,129-135 The
might suffice to correct the problem. It should be reported improved results in the latter reports were
remembered that the serum bilirubin concentration undoubtedly due to the expertise gained at these
rises during the first few days of a normal newborn centers and to the use of ultrasound for guiding the
infant’s life. This rise is due mainly to the deficiency at needle in the peritoneal cavity. In Winnipeg, Canada,
birth of glucoronyl transferase, a liver enzyme respon- for instance, survival rates increased from 70% in
sible for conjugating bilirubin with glucuronyl trans- the period 1970–1978 to 92% during the period
ferase (a liver enzyme responsible for conjugating 1980–1982.11,13,37 Similarly, the survival rate for
bilirubin with glucuronic acid) to form a water-soluble hydropic fetuses in the two periods improved from
compound, bilirubin diglucuronide. This leads to the 50% to 75%. Total fetal fatality rates dues to IUT have
commonly encountered physiologic jaundice, which is dropped to less than 5% at centers who perform this
discussed more fully later in the section on ABO procedure regularly.
HDFN. Bilirubin levels are also affected greatly by Because of erratic adsorption, especially in
prematurity. Hsia and coworkers123 found that in 12 hydropic fetuses, intravascular transfusion has largely
normal full-term infants, peak bilirubin was on average replaced the intraperitoneal approach.39 In an attempt
7.1 mg/dL and was in all cases less than 13.5 mg/dL. to save two hydropic fetuses, Rodeck and associ-
In contrast, the mean bilirubin concentration in 15 ates132,133 used fetoscopy to transfuse the fetuses
premature infants (birthweights below 2250 g) was directly into umbilical vessels, at 23 and 25 weeks’
11.2 mg/dL, and in five infants the concentration gestation, respectively; one baby was delivered at
exceeded 15 mg/dL. The amount of albumin present in 30 weeks and survived. In 1984, Rodeck and associ-
the plasma also has an effect on bilirubin. Plasma ates133 reported using the technique on 25 severely
bilirubin is normally bound to albumin, and the risk of affected fetuses (including 15 with hydrops); the
damage to the nervous system depends on the amount neonatal survival rate was 90%. In 1986, Grannum
of bilirubin that is unbound to albumin. Because of this, and colleagues134 performed in utero exchange trans-
some workers have recommended infusing albumin to fusions on four hydropic fetuses at 24–30 weeks’
babies with HDFN.2 gestation; three of the four babies survived. Perinatal
Table 13-9 shows recommendations of the Scientific survival rates for severely affected fetuses after
Coordinating Committee of the AABB for tests at intravascular IUT are now reported in excess of
delivery.28 80%.13,39
Hemolytic Disease of the Fetus and Newborn 533

TABLE 13-9. RECOMMENDED TESTING AT DELIVERY28

Indication

Maternal Blood
ABO/D To obtain concordant results of tests on two samples, or if pretransfusion tests requested
Antibody detection When pretransfusion tests requested
Antibody identification First detection of alloantibody, D– panel should be used if Rh Ig given during pregnancy
Titration studies Not indicated
FMH testing All D– women who deliver a D+ infant
Testing to diagnose HDFN ABO/D typing and tests for unexpected antibodies if not done during the admission for delivery
Test maternal serum against paternal RBCs if there are no unexpected antibodies found by
routine reagent screen RBCs and no fetomaternal ABO incompatibility
Cord or Infant Blood
Infants born to D– women D status, including test for weak D
Infants born to women with potentially
significant antibodies ABO, D typing, and DAT
No maternal alloimmunization; infant with
clinical signs and symptoms of HDFN ABO/D typing and DAT
If fetomaternal ABO incompatibility exists, infant serum should be tested for IgG anti-A and/or -B
If no fetal maternal ABO incompatibility exists, maternal serum (see above) or infant eluate
should be tested against paternal RBCs

Currently, the decision to perform IUT is usually bilirubin levels.134 The blood used for exchange trans-
based on the amniotic fluid OD450, but as discussed fusion should preferably be less than 48 hours old and
previously, other tests (e.g., fetal hemoglobin, MMA) usually partially “packed.” Most neonatologists find
might also be useful in indicating the need for this CPD-A anticoagulated blood acceptable, but some
treatment. The fetal hematocrit at which to initiate insist on heparinized blood that is less than 12 hours
transfusion is somewhat arbitrary, but ACOG quotes a old. The blood should be ABO-compatible with the
hematocrit of 25% as being a reasonable indication.39 infant and lack the offending antigen (e.g., D-negative
Repeat transfusions are planned for when the fetal blood is used for an infant suffering from Rh(D)-
hematocrit is predicted to be between 20% and 25%. HDFN). As the blood group of the fetus is usually
This may be approximated by assuming a 1% decline unknown before the first IUT, fresh group O blood is
per day or by using one of a number of published usually used for the first IUT.
equations. In fetuses with more severe hemolytic
anemia, few fetal RBCs survive the initial transfusion
interval, whereas transfused adult RBCs survive for a Phototherapy
longer time period. Therefore, the interval between
The irradiation of jaundiced infants with fluorescent
first and second transfusions is usually 7 to 14 days,
lights is currently the most common method for
whereas the interval between subsequent transfusions
treating hyperbilirubinemia, particularly those with
or birth is 21–28 days.39
levels below 20 mg%.136–138 In the United States,
Indication for neonatal exchange transfusion vary
about 2.5% of infants receive phototherapy.137 Some
from center to center but generally, if the cord hemo-
authors have suggested that the technique is over-
globin is less than 12g/dL (hematocrit <45%), or if
used.139,140
cord unconjugated bilirubin is greater than 75
μmol/L, or if the bilirubin rises at a rate of greater
than 10 μmol per hour, most neonatologists would Preterm Delivery
decide to perform an exchange transfusion. The main
reason for performing an exchange transfusion is to Allen141 and Walker and associates142 found that
correct the anemia by removing the baby’s sensitized approximately 50% of all stillbirths due to HDFN
RBCs—which might have a survival time of only 2 to occurred after the 35th week of pregnancy. With the
3 days—and replacing them with donor RBCs with steadily increasing improvement in the case of
normal survival; removal of bilirubin and antibody preterm babies, delivery is now carried out as early as
are other reasons. Subsequent exchanges are based on 28–30 weeks’ gestation, with some centers claiming a
the level of unconjugated bilirubin and its rate of survival rate of greater than 80% after 28 weeks’ ges-
increase, but other factors that influence bilirubin tation. Because of the decreasing incidence of HDFN,
transport and the risk of kernicterus must also be there is decreasing expertise with IUT. There could be
taken into account. These include gestational age, low an increasing trend to deliver babies suffering with
pH, low serum albumin, hypoxia, asphyxia, and infec- HDFN as early as 28–30 weeks rather than to risk
tions, all of which predispose to kernicterus at lower IUT.
534 Immune Hemolytic Anemias

Plasma Exchange RBCs and/or stroma. Yoshida and associates160,161


have reported using the procedure successfully to
The use of plasma exchange on alloimmunzied mothers remove IgG anti-M and P from sera of women who
to ameliorate HDFN is controversial. The first intensive had been unsuccessful in delivering live infants in
plasma exchange was carried out on an Rh-immunized four previous pregnancies, but who after this proce-
woman by Bowman and colleagues143 in 1968. Since dure delivered live babies.
that time, there have been many reports, which were
reviewed by Rock144 in 1984 and by Moise and
Whitecarr145 in 2002. Plasma exchange of three to four Promethazine Hydrochloride (PMT)
liters, up to three or more times a week, might be
required for the duration of a pregnancy with an Treatment
affected fetus. One group claimed that 80% of the There have been several reports of the use of PMT to
mothers showed a reduction in antibody level to at least ameliorate the severity of HDFN.162-166 The largest
half the original level when measured by the Auto- series was reported by Gusdon,165 who treated 72
Analyzer quantitation method.146 Some workers147,148 Rh-immunized women (3.7–6.5 mg PMT/Kg/day
have suggested that smaller volumes of plasma could given orally) for 6 to more than 30 weeks. The total
be removed. For instance, Rubinstein147 removed only number of perinatal deaths was reduced consider-
250 mL of plasma every week for 20 weeks and ably compared with previous pregnancies, with only
decreased antibody levels to 20% of their initial values. three deaths in the treated group; the number of
Nevertheless, plasma exchange should not be under- exchange transfusions was significantly lower, with
taken lightly because the mother is being put to risk as 20 infants requiring 42 exchanges compared with 32
well as the baby; maternal deaths have been reported.149 infants needing 82 exchanges in previously affected
Bowman, quoted in Moise and Whitecarr,145 recom- pregnancies. In contrast, Charles and Blumenthal166
mends carrying out plasma exchange starting at 12–14 did not find any ameliorating effects of PMT in 21
weeks’ gestation if the woman has a homozygous Rh-immunized women; the low dosage used in this
husband, a history of hydropic stillbirths before study has been criticized by others. Scott and
30 weeks’ gestation, and a quantitative anti-D level coworkers167 tried combining PMT treatment with
greater than 2.5 mg/mL. He believes that plasma plasma exchange; Rock144 warns that this combina-
exchange is indicated in very severe Rh disease with tion of immuosuppression and plasma exchange
high levels of anti-D, as these levels can be reduced up could increase the risks of both maternal and neona-
to 80%, if only transiently, by intense plasma exchange. tal infection during pregnancy.
For other Rh-immunized women, he feels the proce-
dure is too costly to be justified. Kemp150 used two-liter
exchanges at least twice a week and showed a live Erythrocyte Membrane Oral
birthrate of 66% for 156 patients who had antibody Treatment (EMOT)
levels greater than 10 IU. If the antibody level cannot be In a letter to Lancet in 1979, Bierme and associates168
lowered sufficiently by plasma exchange, this author’s reported treating seven Rh-immunized women with
center transfuses the fetus using fetoscopy as early as daily treatments of stroma prepared from Rh-
20 weeks’ gestation. Robinson and Tovey151 showed positive erythrocytes. The stromal extract was taken
that use of plasma exchange in their treatment regimen orally in the form of a capsule. All of these women
changed the live birthrate in 14 women from a predicted had previously had stillbirths or intrauterine deaths.
value of 38% to an actual value of 75%. While receiving EMOT, their Rh antibodies did not
rise in titer, and all six of the Rh-positive babies were
Intravenous Immunoglobulin (IVIG) born alive at about 35 weeks’ gestation. When treat-
ment was stopped after delivery, a rapid rise in
IVIG has been used successfully in the antenatal treat- Rh antibody titer occurred in three of the women.
ment of HDFN.145,151-157 Moise’s group uses a combina- In 1982, Bierme and colleagues169 reported on 10
tion of IVIG and plasmapheresis for women who have women who had received EMOT over a 9-year
had a previous fetal loss before 20 weeks’ gestation.145 period. PMT was given in addition to EMOT in 10
of the 11 pregnancies. The women received 2 g of
Specific Immunoadsorption erythrocyte stromal preparation starting at approx-
imately 10 weeks’ gestation. In the group that
Some workers have tried to remove antibody by received EMOT, only 9 of the 11 pregnancies had an
adsorbing the antibody onto allogenic RBCs contain- Rh incompatibility. In these nine pregnancies, the Rh
ing the reactive antigen and then reinfusing the antibody level remained constant and was always
plasma. Tilz and coworkers158 were the first to try this lower than in previous pregnancies. In one case, IUT
with anti-D and Rh(D)-positive RBCs. Robinson159 was performed at 31 weeks’ gestation. All nine
reported initial success with a similar technique but babies were born alive. The control group showed
subsequently found an increase in antibody titer, pre- increases in antibody titer after 30 weeks, and multi-
sumably due to infusion of contaminating Rh-positive ple IUTs were necessary.
Hemolytic Disease of the Fetus and Newborn 535

Other workers have not been so successful with Serological Findings in ABO HDFN
this form of treatment. Barnes and coworkers170
reported on the use of EMOT in 18 nonsensitized MOTHER
Rh(D)-negative male volunteers injected with Rh- Usually, the mother is group O and the baby is A, B, or
positive RBCs and in six Rh-sensitized women. AB, but sometimes the mother is group A or B.2 The
Sixty-one percent (11/18) of the males who received mother’s serum must contain IgG anti-A, anti-B,
EMOT (prepared from either Rh-positive or Rh- and/or anti-A,B. The IgG antibody often is of high
negative RBCs) before intravenous challenge with titer. Voak and Bowley179 found that 66% and 90% of
Rh-positive RBCs produced detectable antibodies; of sera from mothers delivering babies with HDFN due
these 11, six received Rh-negative EMOT, and five to anti-A and anti-B, respectively, contain IgG anti-
received Rh-positive EMOT. Seventy-two (13/18) of bodies with an IAT titer greater than 256. Brouwers
control males who had received no prior EMOT pro- and colleagues180 found that 100% of sera from group
duced Rh antibodies after challenge with Rh-positive O mothers who previously had delivered a group A or
RBCs. Three of six presensitized women who group B baby contained IgG2; 97% of sera contained
received Rh-positive or Rh-negative EMOT for IgG1, 41% contained IgG4, and 38% contained IgG3
4 weeks but without intravenous challenge increased anti-A/B. IgG2 anti-A/B were usually of higher titer
their anti-D levels, which peaked at 11–18 weeks; than anti-A/B of other subclasses. Although the titer
two had received Rh-positive EMOT, and one had and subclass of the antibodies show some correlation
received Rh negative EMOT.170 with the presence or severity of HDFN, they are of
little practical value, especially in antenatal testing.

HDFN ASSOCIATED WITH


ABO ANTIBODIES INFANT
DAT. There is a large range in the reported inci-
In a white population, at least 15% of babies are at dence of positive DATs associated with ABO HDFN.
risk for HDFN due to ABO antibodies.2 Group A or B The low incidence of positive DATs in early reports
infants born to group O mothers are those at greatest probably related to the insensitivity of the AGT
risk, as group O mothers have higher levels of IgG methods used; more recent data report the incidence
anti-A, -B, -A,B in their plasma compared with group as 20%–82%.181-186 One major problem in relating to
A or B mothers. Although many infants are at risk, the incidence quoted in the literature is the authors’
and approximately 5% of all births show some sign of definition of ABO HDFN. Some data relate to positive
ABO HDFN, clinically obvious HDFN is rare.2 ABO- DATs in infants who are ABO incompatible with their
incompatible infants tend to have lower hemoglobin mother, other data relate to infants who have labora-
values and more jaundice (usually slight) than ABO- tory signs of ABO HDFN, and still other data relate to
compatible infants, but the ABO-incompatible infants infants who have evidence of a hemolytic anemia due
rarely need treatment of any kind.2 ABO-incompati- to anti-A or anti-B. The latter is the definition that
ble babies often have positive DATs and/or anti-A,B should be used, but it is often not clear in the publica-
can be eluted from their RBCs, but these findings do tions. One other way of expressing the data is to apply
not equate with a clinical problem most of the time. it to infants who have to be treated for HDFN. Issitt
The need for exchange transfusion is rare (approxi- and Anstee9 reported that infants who have to be
mately 1:1000 to 1:5000 of all births).2 There are sug- treated (transfused) for ABO HDFN always have a
gestions in the literature that ABO HDFN could be positive DAT; this also has been the experience of one
more severe in Africans, African-Americans, Chinese, of the authors (Garratty, unpublished observations).
and Arabs.171-178 There has been much discussion in the literature
When IgG ABO antibodies cross the placenta, they regarding why the DAT in ABO HDFN is often
encounter A and B antigens in the body fluids and weakly positive or sometimes negative. Suggestions
on many cells other than RBCs. Fetal RBCs have less here ranged from pinocytosis of antigen-antibody
A and B sites than adult RBCs, and the carbohydrate complexes, low number of A and B sites in fetal RBCs,
chains on the membrane are less branched com- and deficiency of branched chains on the fetal RBC
pared with adult RBCs. IgG ABO antibodies rarely membrane.186-189
bind complement to the fetal/cord RBCs (i.e., the Eluates from Infant’s RBCs. One can almost always
RBCs react with anti-IgG but not anticomplement elute anti-A or anti-B from group A or B infants born to
antiglobulin sera). All of the RBC destruction is a group O mother, even if the DAT is negative. Because
macrophage mediated, similar to Rh HDFN. Severe of this, detection of anti-A and anti-B in an eluate has an
anemia is uncommon, and hydrops fetalis has been extremely low predictive value for HDFN. A nonreac-
reported in only two or three cases. 2 Hyper- tive eluate might help exclude ABO antibodies as a
bilirubemia leading to kernicterus is the major cause of the positive DAT and/or jaundice.
problem; this usually reaches its peak 24–48 hours Ukita and coworkers185 showed that 88% and 40%
after the baby’s birth.2 of the IgG anti-A or anti-B eluted from A or B infants
536 Immune Hemolytic Anemias

born to group O mothers were IgG2 and IgG1, respec- lengthening the pretreatment observation period.
tively; none were IgG3 or IgG4. Phototherapy was performed if the bilirubin was
Correlation of Serology with HDFN. It is often greater than or equal to 10 mg% at less than 12 hours
difficult to prove that an infant’s jaundice is due to of age; greater than or equal to 12 mg% at less than
ABO HDFN. If the infant is group A or B and the 18 hours of age; greater than or equal to 14 mg%
mother is group O, it is usually assumed that the ABO before 24 hours of age; and greater than 15 mg% after
antibodies are to blame, and if a transfusion is needed, 24 hours of age. Using the new protocol, only 9% of
group O blood is used. infants needed phototherapy; none needed exchange
Dufour and Monaghan183 found that 252 (38%) of transfusion. If the old protocol had been used, 82% of
665 group A and B infants born to group O mothers the infants would have received phototherapy, and
had laboratory findings compatible with a diagnosis 5% would have had exchange transfusions. The
of ABO HDFN (0.8% of ABO-incompatible infants authors commented that exchange transfusion should
born to group A or B mothers), but only 28 (4%) of the be used only for severely compromised or asphyxi-
665 infants (14% of DAT-positive infants) needed ated infants.190
treatment (17 phototherapy; seven phototherapy plus
exchange transfusion; four exchange transfusion
only). Thirty-one percent (208) of the infants were R E F E R E N C E S
DAT positive; 64% of these infants were jaundiced
(26% had bilirubin >12 mg%). Of the DAT-negative 1. Hadley AG, Soothill P (eds): Alloimmune Disorders of
(eluate reactive) infants, 21% (38) were jaundiced (8% Pregnancy. Cambridge: Cambridge University Press, 2002.
had bilirubin >12 mg%). It should be noted that 37% 2. Mollison PL, Engelfriet CP, Contreras M: Blood Transfusion in
Clinical Medicine, 10th ed. Oxford: Blackwell Scientific, 1997.
of the control group (137 unselected infants) had jaun- 3. Giblett ER: Blood group antibodies causing hemolytic disease
dice (7% had bilirubin >12 mg%). of the newborn. Clin Obstet Gynecol 1964;7:1044–1055.
Desjardins and colleagues182 reported on 1704 4. Polesky HF: Blood group antibodies in prenatal sera. Minn
infants born to group O mothers. Of the 680 ABO- Med 1967;50:601–603.
5. Queenan JT, Smith BD, Haber JM, Jeffrey J, Gadow HC:
incompatible infants, 27 (4%) had clinically significant Irregular antibodies in the obstetric patient. Obstet Gynecol
ABO HDFN. The DAT was positive in 22 (82%) of 1969;34:767–770.
these infants; in four infants (15%), the DAT was neg- 6. Walker RH: Relevancy in the selection of serologic tests for the
ative, but the eluate was reactive. The group A and B obstetric patient. In Garratty G (ed): Hemolytic Disease of the
infants had significantly higher bilirubins and lower Newborn. Arlington, VA: American Association of Blood
Banks, 1984:173–210.
hemoglobins than the group O infants. 7. Kornstad L: Occurrence of anti-D and other irregular blood
Osborn and coworkers190 reported on 476 infants group antibodies in pregnancy. The present situation. In Book
born at the University of California, Los Angeles. of Abstracts, XXI Congress of the International Society of
Forty-seven of the infants (10%, all infants were born Haematology, XIX Congress of the International Society of
Blood Transfusion, 1986:312.
to group O mothers), were DAT positive. Of the DAT- 8. Kornstad L: New cases of irregular blood group antibodies
positive infants, 92% were found not to have clinically other than anti-D in pregnancy. Acta Obstet Gynecol Scand
significant HDFN and had bilirubin levels indistin- 1983;62:431–436.
guishable from controls (12%–14% of both groups had 9. Issitt PD, Anstee DJ: Applied Blood Group Serology, 4th ed.
jaundice). The strength of the DAT was found to be Durham, NC: Montgomery Scientific Publications, 1998.
10. Wenk RE, Goldstein P, Felix JK: Alloimmunization by hr’(c),
unrelated to the need for phototherapy. The authors hemolytic disease of newborns, and perinatal management.
emphasized that no laboratory tests predicted HDFN Obstet Gynecol 1986;67:623–626.
with 100% accuracy and that careful, continuous 11. Bowman JM: Hemolytic disease of the newborn. In Garratty G
monitoring of the bilirubin was the only reliable indi- (ed): Immunobiology of Transfusion Medicine. New York:
Dekker, 1994:553–595.
cator for treatment. The authors developed new, more 12. Wenk RE, Goldstein P, Felix JK: Kell alloimmunization,
conservative criteria for treatment of ABO HDFN. The hemolytic disease of the newborn, and perinatal manage-
old criteria, when the infants was less than 24 hours ment. Obstet Gynecol 1985;66:473–476.
old, were that phototherapy was performed if the 13. Bowman JM: Historical overview: Hemolytic disease of the
infant’s bilirubin was 5–9 mg%; if the bilirubin was fetus and newborn. In Kennedy MS, Wilson S, Kelton JG (eds):
Perinatal transfusion medicine. Arlington, VA: American
greater than 10 mg%, exchange transfusion was Association of Blood Banks, 1990:1–52.
performed instead. When the infant was greater 14. Caine ME, Mueller-Heubach E: Kell sensitization in preg-
24 hours old, if the bilirubin was 14–20 mg%, then nancy. Am J Obstet Gynecol 1986;154:85–90.
phototherapy or exchange transfusion, respectively, 15. Mayne KM, Bowell PJ, Pratt GA: The significance of anti-Kell
sensitization in pregnancy. Clin Lab Haemat 1990;12:379–385.
was performed. Retrospective analysis showed that 16. Bowman JM, Pollock JM, Manning FA, et al: Maternal Kell
DAT-positive, ABO-incompatible infants were often blood group alloimmunization. Obstet Gynecol 1992;79:
treated with phototherapy for 24–48 hours and never 239–244.
developed nonphysiologic jaundice; once treated, 17. McKenna DS, Nagaraja HN, O’Shaughnessy R: Management
bilirubin seldom continued rising, and very few of pregnancies complicated by anti-Kell isoimmunization.
Obstet Gynecol 1999;93:667–673.
infants ever developed bilirubin levels high enough to 18. Grant SR, Kilby MD, Meer L, et al: The outcome of pregnancy
warrant exchange transfusion. The criteria were in Kell alloimmunisation. Br J Obstet Gynaecol 2000;107:
changed to decrease the number of infants treated by 481–485.
Hemolytic Disease of the Fetus and Newborn 537

19. Berkowitz RL, Beyth Y, Sadovsky E: Death in utero due to Kell 43. Nicolaides KH, Fontanarosa M, Gabbe SG, et al: Failure of
sensitization without excessive elevation of the ΔOD450 value ultrasonographic parameters to predict the severity of fetal
in amniotic fluid. Obstet Gynecol 1982;60:746–749. anemia in rhesus isoimmunization. Am J Obstet Gynecol
20. Babinszki A, Lapinski RH, Berkowitz RL: Prognostic factors 1988;158:920–926.
and management in pregnancies complicated with severe Kell 44. Mari G: Noninvasive diagnosis by Doppler ultrasonography
alloimmunization: Experiences of the last 13 years. Am J of fetal anemia due to maternal red-cell alloimmunization.
Perinatol 1998;15:695–701. N Engl J Med 2000;342:9–14.
21. Vaughan JI, Warwick R, Letsky E, et al: Erythropoietic sup- 45. Stefos T, Cosmi E, Detti L, et al: Correction of fetal anemia on
pression in fetal anemia because of Kell alloimmunization. the middle cerebral artery peak systolic velocity. Obstet
Am J Obstet Gynecol 1994;171:247–252. Gynecol 2002;99:211–215.
22. Vaughan JI, Manning M, Warwick RM, et al: Inhibition of ery- 46. Rodeck CH, Nicolaides KH: Ultrasound guided invasive
throid progenitor cells by anti-Kell antibodies in fetal alloim- procedures in obstetrics. Clinics Obstet Gynecol 1983;10:
mune anemia. N Eng J Med 1998;338:798–803. 515–539.
23. Hardy J, Napier JAF: Red cell antibodies detected in antenatal 47. MacKenzie IZ, Guest CM, Bowell PJ: Fetal blood group
tests on rhesus positive women in South and Mid Wales, studies during mid-trimester pregnancy and the management
1948–1978. Br J Obstet Gynaecol 1981;88:91. of severe isoimmunization. Prenatal Diagn 1983;3:41–46.
24. Bowell PJ, Allen DL, Entwistle CC: Blood group antibody 48. MacKenzie IZ: Fetoscopy in the management of hemolytic
screening tests during pregnancy. Br J Obstet Gynaecol disease of the newborn. Plasma Ther Transfus Technol
1986;93:1038–1043. 1984;5:33–42.
25. Bowell PJ, Brown SE, Dike AE, Inskip MJ: The significance of 49. Nicolaides KH, Rodeck CH, Mibashan RS, Kemp JR: Have
anti-c alloimmunisation in pregnancy. Br J Obstet Gynaecol Liley charts outlived their usefulness? Am J Obstet Gynecol
1986;93:1044–1048. 1986;155:90–94.
26. Rothenberg JM, Weirermiller B, Dirig K, et al: Is a third- 50. Daffos F, Capella-Pavlovsky M, Forestier F: A new procedure
trimester antibody screen in Rh+ women necessary? Am J for fetal blood sampling in utero: Preliminary results of
Manag Care 1999;5:1145–1150. 53 cases. Am J Obstet Gynecol 1983;146:985.
27. Heddle NM, Klama L, Frassetto R, et al: A retrospective study 51. Daffos F, Capella-Pavlovsky M, Forestier F: Fetal blood sam-
to determine the risk of red cell alloimmunization and trans- pling during pregnancy with the use of a needle guided by
fusion during pregnancy. Transfusion 1993;23:217–220. ultrasound: A study of 606 consecutive cases. Am J Obstet
28. Judd WJ: Practice guidelines for prenatal and perinatal Gynecol 1985;153:655–60.
immunohematology, revisited. Transfusion 2001;41:1445–1452. 52. Hobbins JC, Grannum PA, Romero R, Reece EA, Mahoney MJ:
29. Hughes-Jones NC: The estimation of the concentration and Percutaneous umbilical blood sampling. Am J Obstet Gynecol
equilibrium constant of anti-D. Immunology 1967;12: 1985;152:1–6.
565–571. 53. Philip J, Brandt NJ, Fernandes A, Freiesleben E, Trolle D: ABO
30. Contreras M: Methods for quantitation of anti-D. Plasma Ther and Rh phenotyping of foetal blood obtained by foetoscopy.
Transfus Technol 1984;5:65–71. Clin Genet 1978;14:324–329.
31. Gall SA, Miller JM, Jr. Rh isoimmunization: A 24 year experi- 54. Habibi B, Bretagne M, Bretagne Y, Forestier F, Daffos F: Blood
ence at Duke University Medical Center. Am J Obstet Gynecol group antigens on fetal red cells obtained by umbilical vein
1981;140:902–908. puncture under ultrasound guidance: A rapid hemagglutina-
32. Bowell P, Wainscoat JS, Peto TEA, Gunson HH: Maternal anti- tion test to check for contamination with maternal blood.
D concentrations and outcome in rhesus haemolytic disease of Pediatr Res 1986;20:1082–1084.
the newborn. Br Med J 1982;285:327–329. 55. Gemke RJBJ, Kanhai HHH, Overbeeke MAM, et al: ABO and
33. Bowell PJ, Wainscoat JS, Peto TEA: AutoAnalyzer anti-D Rhesus phenotyping of fetal erythrocytes in the first trimester
measurement and outcome of rhesus-sensitized pregnancies: of pregnancy. Brit J Haemat 1986;64:689–697.
Serum analysis. Plasma Ther Transfus Technol 1984;5:81–85. 56. Kanhai HH, Gravenhorst JB, Gemke RJ, Overbeeke MA,
34. Nicolaides KH, Rodeck CH: Maternal serum anti-D antibody Bernini LF, Beverstock GC: Fetal blood group determination
concentration and assessment of rhesus isoimmunisation. Br in first trimester pregnancy for the management of severe
Med J 1992;304:1155–1156. immunization. Am J Obstet Gynecol 1987;156:120–123.
35. Bevis DCA: Composition of liquor amnii in haemolytic 57. Rodesch F, Lambermount M, Donner C, et al: Chorionic
disease of the newborn. Lancet 1950;ii:443. biopsy in management of severe Kell alloimmunization. Am J
36. Liley AW: Liquor amnii analysis in management of pregnancy Obstet Gynecol 1987;156:124–125.
complicated by rhesus immunization. Am J Obstet Gynecol 58. Jackson LG, Zachary JM, Fowler SE, et al: A randomized
1961;82:1359. comparison of transcervical and transabdominal chorionic-
37. Bowman JM: Management of Rh-isoimmunization. Obstet villus sampling. The U.S. National Institute of Child Health
Gynecol 1978;52:1. and Human Development Chorionic-Villus Sampling and
38. Queenan JT, Tomai TP, Ural SH, et al: Deviation in amniotic Amniocentesis Study Group. N Engl J Med 1992;327:594–598.
fluid optical density at a wavelength of 450 nm in Rh- 59. Avent ND: Antenatal genotyping of the blood groups of the
immunized pregnancies from 14 to 40 weeks gestation: A fetus. Vox Sang 1998;74(S2):365–374.
proposal for clinical management. Am J Obstet Gynecol 60. Avent N: Fetal genotyping. In Hadley AG, Soothill P (eds):
1993;168:1370–1376. Alloimmune Disorders of Pregnancy. Cambridge: Cambridge
39. The American College of Obstetricians and Gynecologists. University Press, 2002:121–139.
Management of isoimmunization in pregnancy. Washington, 61. Allen RW, Ward S, Harris R: Prenatal genotyping for the RhD
DC: American College of Obstetricians and Gynecologists, blood group antigen: Considerations in developing an accu-
1996 (ACOG Technical Bulletin. Number 227:1–7). rate test. Genet Test 2000;4:377–381.
40. Moise KJ Jr: Changing trends in the management of red blood 62. Garratty G: Predicting the clinical significance of red cell anti-
cell alloimmunization in pregnancy. Arch Pathol Lab Med bodies with in vitro cellular assays. Transfus Med Rev 1990;
1994;118:421–428. 4:297–312.
41. Weiner S, Bolognese RJ, Librizzi RJ: Ultrasound in the evalua- 63. Engelfriet CP, Ouwehand WH: ADCC and other cellular bio-
tion and management of the isoimmunized pregnancy. J Clin assays for predicting the clinical significance of red cell alloan-
Ultrasound 1981;9:315–323. tibodies. In Contreras M (ed): Blood transfusion: The Impact
42. Abdel-Fattah, Soothill P: Assessing the severity of haemolytic of New Technologies. Baillière’s Clinical Haematology.
disease of the fetus and newborn: Clinical aspects. In Hadley London: Baillière, Tindall, 1990:321–339.
AG, Soothill P, eds. Alloimmune disorders of pregnancy. 64. Zupanska B: Cellular immunoassays and their use for pre-
Cambridge: Cambridge University Press, 2002:153–172. dicting the clinical significance of antibodies. In Garratty G
538 Immune Hemolytic Anemias

(ed): Immunobiology of Transfusion Medicine. New York: 84. Nance SJ, Arndt PA, Garratty G: Correlation of IgG subclass
Dekker, 1994:465–491. with the severity of hemolytic disease of the newborn.
65. Engelfriet CP, Reesink HW: Laboratory procedures for the Transfusion 1990;30:381–382.
prediction of the severity of haemolytic disease of the 85. Zupanska B, Brojer E, Richards Y, et al: Serological and
newborn. Vox Sang 1995;69:61–69. immunological characteristics of maternal anti-Rh(D) anti-
66. Zupanska B, Lenkiewicz B, Michalewska B, et al: The ability bodies in predicting the severity of haemolytic disease of the
of cellular assays to predict the necessity for cordocenteses in newborn. Vox Sang 1989;56:247–253.
pregnancies at risk of haemolytic disease of the newborn.Vox 86. Pollock JM, Bowman JM: Anti-Rh(D) IgG subclass and sever-
Sang 2001;80:234–235. ity of Rh hemolytic disease of the newborn. Vox Sang
67. Urbaniak SJ, Greiss MA, Crawford RJ, et al: Prediction of the 1990;59:176–179.
outcome of rhesus haemolytic disease of the newborn. 87. Alie-Duran SJ, Dugoujon J-M, Fournie A: Gm typing, IgG sub-
Additional information using an ADCC assay. Vox Sang classes of anti-Rh(D) and severity of hemolytic disease of the
1984;46:323–329. newborn. Vox Sang 1992;62:127–128.
68. Oepkes D, van Kamp IL, Simon MJG, et al: Clinical value of 88. Iyer YS, Kulkarni SV, Gupte SC: Distribution of IgG subtypes
an antibody-dependent cell-mediated cytotoxicity assay in the in maternal anti-D sera and their prognostic value in Rh
management of RhD alloimmunization. Am J Obstet Gynecol haemolytic disease of the new-born. Acta Haematol 1992;88:
2001;184:1015–1020. 78–81.
69. Nance S, Nelson J, O’Neill P, et al: Correlation of monocyte 89. Lambin P, Debbia M, Puillandre P, et al: Anti-D IgG1 and IgG3
monolayer assays, maternal antibody titers, and clinical in maternal serum and on the red blood cells of infants suffer-
course in hemolytic disease of the newborn (HDFN) ing from hemolytic disease of the newborn: Relationship with
[abstract]. Transfusion 1984;24:415. the severity of the disease. Transfusion 2002;42:1537–1546.
70. Nance SJ, Nelson J, Horenstein J, et al: Monocyte monolayer 90. Levine P: Serological factors as possible causes in sponta-
assay: An efficient noninvasive technique for predicting the neous abortions. J Heredity 1943;34:71–80.
severity of hemolytic disease of the newborn. Am J Clin 91. Race RR, Sanger R: Blood Groups in Man. Oxford: Blackwell
Pathol 1989;92:89–92. Scientific, 1950:234–236.
71. Sacks DA, Nance SJ, Garratty G, et al: Monocyte monolayer 92. Nevanlinna HR, Vainio T: The influence of mother-child ABO
assay as a predictor of severity of hemolytic disease of the incompatibility on Rh immunisation. Vox Sang 1956;1:26.
fetus and newborn. Am J Perinatol 1993;10:428–431. 93. Finn R: Erythroblastosis. Lancet 1960;1:526.
72. Zupanska B, Brojer E, Richards Y, et al: Serological and 94. Finn R, Harper DT, Stallings SA, et al: Transplacental hemor-
immunological characteristics of maternal anti-Rh(D) anti- rhage. Transfusion 1963;3:114–124.
bodies in predicting the severity of haemolytic disease of the 95. Finn R, Clarke CA, Donohoe WTA, et al: Experimental studies
newborn. Vox Sang 1989;56:247–253. on the prevention of Rh haemolytic disease. Br Med J 1961;I:
73. Zupanska B: Cellular immunoassays and their use for pre- 1486–1490.
dicting the clinical significance of antibodies. In Garratty G 96. Clarke CA, Donohoe WTA, McConnell RB, et al: Further
(ed): Immunobiology of transfusion medicine. New York: experimental studies in the prevention of Rh-haemolytic
Dekker, 1994:465–491. disease. Br Med J 1963;1:979–984.
74. Larson PJ, Thorp JM Jr, Miller RC, et al: The monocyte 97. Stern K, Davidsohn I, Masaitis L: Experimental studies on Rh
monolayer assay: A noninvasive technique for predicting the immunization. Am J Clin Pathol 1956;26:833–843.
severity of in utero haemolysis. Am J Perinatol 1995;12: 98. Stern K, Goodman HS, Berger M: Experimental iso-
157–160. immunization to hemo-antigens in man. J Immunol 1961;
75. Moise KJ, Perkins JT, Sosler SD, et al: The predictive value of 87:189–198.
maternal serum testing for detection of fetal anaemia in red 99. Clarke CA, Donohoe WTA, McConnell RB, et al: Further
blood cell alloimmunization. Am J Obstet Gynecol 1995;172: experimental studies on the prevention of Rh haemolytic
1003–1009. disease. Br Med J 1963;I:979–984.
76. Hadley AG, Kumpel BM: The role of Rh antibodies in 100. Smith T: Active immunity produced by so-called balanced or
haemolytic disease of the newborn. Bailliere Clin Haematol neutral mixtures of diptheria toxin and antitoxin. J Exp Med
1993;6:423–444. 1909;11:241–256.
77. Frankowska K, Gorska B: IgG subclasses of anti-Rh antibodies 101. Freda VJ, Gorman JG: Current concepts: Antepartum man-
in pregnant women. Arch Immunol et Ther Exper 1978; agement of Rh hemolytic disease. Bull Sloane Hosp Women
26:1095–1100. 1962;8:147–158.
78. Ouwehand WH, Engesser L, Huiskes E, et al: Differences in 102. Freda VJ, Gorman JG, Pollack W: Successful prevention of
efficacy of IgG1 and IgG3 antibodies in the destruction of red experimental Rh sensitisation in man with an anti-Rh
cells. In Ouwehand WH (ed): The activity of IgG1 and IgG3 gammaglobulin antibody preparation. Transfusion 1964;4:
antibodies in immune mediated destruction of red cells 26–32.
[thesis]. Amsterdam: Rodopi, 1984:44–63. 103. Combined study. Prevention of Rh haemolytic disease:
79. Parinaud J, Blanc M, Grandjean H, et al: IgG subclasses and Results of the clinical trial. A combined study from centres in
Gm allotypes of anti-D antibodies during pregnancy: England and Baltimore. Br Med J 1966;ii:907–914.
Correlation with the gravity of the fetal disease. Am J Obstet 104. Pollack W, Gorman JG, Freda VJ, Ascari WQ, Allen AE, Baker
Gynecol 1985;151:1111–1115. WJ: Results of clinical trials of RhoGAM in women.
80. Nance S, Arndt P, Nelson J, et al: Correlation of IgG subclass Transfusion 1968;8:151–153.
with the severity of hemolytic disease of the newborn 105. Pollack W, Kochesky RJ: The importance of antibody concen-
[abstract]. Transfusion 1989;29:48S. tration, binding constant, and heterogeneity in the suppres-
81. Schanfield MS: Human immunoglobulin (IgG) subclasses and sion of immunity to the Rh factor. Int Arch Allergy
their biological properties. In Dawson RB (ed): Blood Bank 1970;38:320–336.
Immunology. Washington, DC: American Association of 106. Sebring ES, Polesky HF: Fetomaternal hemorrhage: Incidence,
Blood Banks, 1977:97–112. risk factors, time of occurrence, and clinical effects.
82. Cregut R, Pinon F, Brossard Y: Sous-classes d’immunoglobu- Transfusion 1990;30:344–357.
lines anti-Rh(D) et maladie hemolytique du nouveau-ne. 107. Zipursky A, Israels LG: The pathogenesis and prevention of
Nouv Presse Med 1973;29:1947–1949. Rh immunization. Canad Med Assoc J 1967;97:1245.
83. Taslimi MM, Sibai BM, Mason JM, et al: Immunoglobulin G 108. Bowman JM, Chown B, Lewis M, et al: Rh immunization
subclasses and isoimmunized pregnancy outcome. Am J during pregnancy. Antenatal prophylaxis. Canad Med Assoc J
Obstet Gynecol 1986;154:1327–1332. 1978;118:623.
Hemolytic Disease of the Fetus and Newborn 539

109. McMaster Conference on Prevention of Rh Immunization. 135. Berkowitz RL, Chitkara U, Goldberg JD, Wilkins I, Chervenak
Vox Sang 1979;36:50–64. FA: Intravascular transfusion in utero: the percutaneous
110. Visscher RD, Visscher HC: Do Rh negative women with an approach. Am J Obstet Gynecol 1986;154:622–623.
early spontaneous abortion need Rh immune prophylaxis? 136. Speidel BD: Pediatric management of the severely affected
Am J Obstet Gynecol 1972;113:158–162. rhesus baby. Plasma Ther Transfus Technol 1984;5:43–46.
111. Bowman JH: The prevention of Rh immunization. Transfus 137. Brown AK, McDonagh AF: Phototherapy for neonatal hyper-
Med Rev 1988;2:129–150. bilirubinaemia: Efficiency, mechanism and toxicity. Adv
112. Bowman JH: Controversies in Rh prophylaxis. In Garratty G Paediatr 1980;27:341–389.
(ed): Hemolytic disease of the newborn. Arlington, VA: 138. Mathew PM, Wharton BM: Investigation and management of
American Association of Blood Banks, 1984:67–85. neonatal jaundice: A problem oriented case record. Arch Dis
113. Urbaniak SJ: Rh(D) haemolytic disease of the newborn: The Child 1981;56:949–953.
changing scene. Br Med J 1985;291:4–6. 139. Lewis HM, Campbell RHA, Hambleton G: Use or abuse of
114. Tovey GH: Should anti-D immunoglobulin be given antena- phototherapy for physiological jaundice of newborn infants.
tally? Lancet 1980;ii:466–468. Lancet 1982;ii:408–410.
115. Nusbacher J, Bove JR: Rh immunoprophylaxis: is antepartum 140. Hein HA: Why do we keep using phototherapy in healthy
therapy desirable? N Eng J Med 1980;303:935–937. newborns? Pediatrics 1984;73:881–882.
116. Tovey LAD, Taverner JM: A case for the antenatal adminis- 141. Allen FH Jr: Induction of labor in the management of ery-
tration of anti-D immunoglobulin to primigravidae. Lancet throblastosis fetalis. Quart Rev Pediat 1957;12:1.
1981;i:878–881. 142. Walker W, Murray S, Russell JK: Stillbirth due to haemolytic
117. Mollison PL: Some aspects of Rh hemolytic disease and its disease of the newborn. J Obstet Gynaec Br Emp 1957;44:573.
prevention. In Garratty G (ed): Hemolytic disease of the 143. Bowman JM, Peddle LJ, Anderson C: Plasmapheresis in
newborn. Arlington, VA: American Association of Blood severe Rh iso-immunization. Vox Sang 1968;15:272–277.
Banks, 1984:1–32. 144. Rock G: Amelioration of Rh disease. In Garratty G (ed):
118. Brouwers HAA, Overbeeke MAM, Huiskes E, et al: Comple- Hemolytic Disease of the Newborn. Arlington, VA: American
ment is not activated in ABO-haemolytic disease of the Association of Blood Banks, 1984:119–143.
newborn. Br J Haematol 1988;68:363–366. 145. Moise KJ Jr, Whitecarr PW: Antenatal therapy for haemolytic
119. Vescio LAC, Castro RA: Complement is not activated in disease of the fetus and newborn. In Hadley AG, Soothill P
ABO-hemolytic disease of the newborn: further support. Vox (eds): Alloimmune Disorders of Pregnancy. Cambridge:
Sang 1990;58:231. Cambridge University Press, 2002:73–201.
120. Pochedly C, Palladino N: Etiology of late anemia of hemolytic 146. Fraser ID, Bennett MO, Bothamley JE, Airth GR: Intensive
disease of the newborn. Clin Med 1971;78:30–34. antenatal plasmapheresis in severe rhesus isoimmunisation.
121. Hughes-Jones NC, Hughes MIJ, Walker W: The amount of Lancet 1976;1:6–9.
anti-D on red cells in haemolytic disease of the newborn. Vox 147. Rubinstein P: Repeated small volume plasmapheresis in the
Sang 1967;12:279–285. management of hemolytic disease of the newborn. In
122. Garratty G, Nance S: Correlation between in vivo hemolysis Frigoletto SD, Jewett JF, Konugres AA (eds): Rh Hemolytic
and the amount of red cell bound IgG measured by flow Disease, New Strategy for Eradication. Boston: GK Hall and
cytometry. Transfusion 1990;30:617–621. Co, 1981:211–220.
123. Hsia DY-Y, Allen FH, Gellis SS, et al: Erythroblastosis fetalis. 148. Eernisse JG, Gravenhorst JB: 2. Prevention of severe
VIII. Studies of serum bilirubin in relation to kernicterus. haemolytic disease of the newborn by weekly small-volume
N Engl J Med 1952;247:668. plasmapheresis during pregnancy. In Sibinga CTS, Das PC,
124. Allen FH Jr, Diamond LK, Vaughan VC III: Erythroblastosis Forfar JO (eds): Paediatrics and Blood Transfusion. Boston:
fetalis. VI. Prevention of kernicterus. Am J Dis Child Martinus Nijhoff, 1982:164–170.
1905;80:779–791. 149. Huestis D: Mortality in therapeutic haemapheresis. Lancet
125. Allen FH Jr, Diamond LK, Jones AR: Erythroblastosis fetalis. 1983;i:1043.
IX. The problems of stillbirth. N Eng J Med 1954;251: 150. Kemp JR: Plasmapheresis: The Lewisham experience. Plas
453–459. Ther 1984;5:21–22.
126. Liley AW: Intrauterine transfusion of foetus in haemolytic 151. Robinson AE, Tovey LAD: Intensive plasma exchange in the
disease. Br Med J 1963;ii:1107–1109. management of severe Rh disease. Br J Hematol 1980;45:
127. Robertson EG, Brown A, Ellis MI, Walker W: Intrauterine 621–631.
transfusion in the management of severe Rhesus isoimmu- 152. Marguiles M, Voto LS, Mathet E: High-dose intravenous IgG
nization. Brit J Obstet Gynecol 1976;83:694–697. for the treatment of severe Rhesus alloimmunization. Vox
128. Palmer A, Gordon RR: A critical review of intrauterine fetal Sang 1991;61:181–189.
transfusion. Brit J Obstet Gynecol 1976;83:688–693. 153. Voto LS, Mathet ER, Zapaterio JL, et al: High-dose gamma-
129. Hamilton EG: Intrauterine transfusion. Am J Obstet Gynecol globulin (IVIG) followed by intrauterine transfusions (IUT): A
1977;50:255–260. new alternative for the treatment of severe fetal hemolytic
130. Frigoletto FD Jr, Umansky I, Birnholz J, et al: Intrauterine fetal disease. J Perinat Med 1997;25:85–88.
transfusion in 365 fetuses during fifteen years. Am J Obstet 154. Dooren MC, van Kamp IL, Scherpenisse JW, et al: No
Gynecol 1981;139:781–790. beneficial effect of low-dose fetal intravenous gammaglobulin
131. Buscaglia M, Ferrazzi E, Zuliani G, Caccamo ML, Pardi G: administratin in combination with intravascular transfusions
Ultrasound contributions to the management of the severely in severe Rh D haemolytic disease. Vox Sang 1994;66:253–257.
isoimmunized fetus. J Perinat Med 1986;14:51–58. 155. Rubo J, Albrecht K, Lasch P, et al: High-dose intravenous
132. Rodeck CH, Holman CA, Karnicki J, Kemp JR, Whitmore DN, immune globulin therapy for hyperbilirubinemia caused by
Austin MA: Direct intravascular fetal blood transfusion by Rh hemolytic disease. J Pediatr 1992;121:93–97.
fetoscopy in severe Rhesus isoimmunisation. Lancet 156. Alonso JG, Decaro J, Marrero A, et al: Repeated direct fetal
1981;i:625–627. intravascular high-dose immunoglobulin therapy for the
133. Rodeck CH, Nicolaides KH, Warsof SL, Fysh WJ, Gamsu HR, treatment of Rh hemolytic disease. J Perinat Med
Kemp JR: The management of severe rhesus isoimmunization 1994;22:415–419.
by fetoscopic intravascular transfusions. Am J Obstet Gynecol 157. Gottvall T, Selbing A: Alloimmunization during pregnancy
1984;150:769–774. treated with high dose intravenous immunoglobulin. Effects
134. Grannum PA, Copel JA, Plaxe SC, et al: In utero exchange on fetal hemoglobin concentration and anti-D concentrations
transfusion by direct intravascular injection in severe ery- in the mother and fetus. Acta Obstet Gynecol Scand
throblastosis fetalis. N Engl J Med 1986;314:1431–1434. 1995;74:777–783.
540 Immune Hemolytic Anemias

158. Tilz GP, Weiss PAM, Teubl I, Lanzer G, Vollmann H: 175. Chan-Shu SY, Blair O: ABO hemolytic disease of the newborn.
Succeessful plasma exchange in rhesus incompatibility. Am J Clin Pathol 1979;71:677–679.
Lancet 1977;2:203. 176. Vos GH, Adhikari M, Coovadia HM: A study of ABO incom-
159. Robinson AE: Unsuccessful use of absorbed autologous patibility and neonatal jaundice in Black South African
plasma in Rh incompatible pregnancy. N Eng J Med newborn infants. Transfusion 1981;21:744–749.
1981;305:1346. 177. Feng CS, Wan CP, Lau J, et al: Incidence of ABO haemolytic
160. Yoshida Y, Yoshida H, Tatsumi K, et al: Successful antibody disease of the newborn in a group of Hong Kong babies with
elimination in severe M-incompatible pregnancy. N Eng J severe neonatal jaundice. Paediatr Child Health
Med 1981;305:460–461. 1990;26:155–157.
161. Yoshida H, Ito K, Emi N, Kanzaki H, Matsuura S: A new 178. Lin M, Broadberry RE: ABO hemolytic disease of the newborn
therapeutic antibody removal method using antigen-positive is more severe in Taiwan than in White populations. Vox Sang
red cells. Vox Sang 1984;47:216–223. 1995;68:136.
162. Gusdon JP Jr, Witherow C: Possible ameliorating effects of 179. Voak D, Bowley CC: A detailed serological study on the pre-
erythroblastosis by promethazine hydrochloride. Am J Obstet diction and diagnosis of ABO haemolytic disease of the
Gynecol 1973;117:1101–1108. newborn (ABO HD). Vox Sang 1969;17:321–348.
163. Rubinstein A, Eidelman AI, Melamed J, Gartner LM, Kandall 180. Brouwers HAA, Overbeeke MAM, Gemke RJBJ, et al:
S, Schulman H: Possible effect of maternal promethazine Sensitive methods for determining subclasses of IgG anti-A
therapy on neonatal immunologic functions. J Pediatr and anti-B in sera of blood–group-O women with a blood-
1976;89:136–138. group-A or -B child. Br J Haematol 1987;66:267–270.
164. Stenchever MA: Promethazine hydrochloride: use in patients 181. Orzalesi M, Gloria F, Lucarelli P, et al: ABO system incompa-
with Rh isoimmunization. Am J Obstet Gynecol 1978;130: tibility: Relationship between direct Coombs test positivity
665–668. and neonatal jaundice. Pediatr 1973;51:288–289.
165. Gusdon JP Jr: The treatment of erythroblastosis with promet- 182. Desjardins L, Chintu C, Zipursky A: The spectrum of ABO
hazine hydrochloride. J Reprod Med 1981;26:454–458. hemolytic disease of the newborn infant. J Pediatr 1979;95:
166. Charles AG, Blumenthal LS: Promethazine hydrochloride 447–449.
therapy in severely Rh sensitized pregnancies. Obstet Gynecol 183. Dufour DR, Monaghan WP: ABO hemolytic disease of the
1982;60:627–630. newborn. Am J Clin Pathol 1980;73:369–373.
167. Scott JR, Anstall HB, Rote NS, Kochenour NK, Beeson JH: 184. Levine DH, Belton H, Meyer P: Newborn screening for ABO
Effect of plasma exchange and immunosuppression on hemolytic disease. Clin Pediatr 1985;24:391–394.
Rho(D), viral, and bacterial antibody titers in Rh immunized 185. Ukita M, Takahashi A, Nunotani T, et al: IgG subclasses of
women. Am J Reprod Immunol 1982;2:46–49. anti-A and anti-B antibodies bound to the cord red cells in
168. Bierme SJ, Blanc M, Abbal M, Fournie A: Oral Rh treatment ABO incompatible pregnancies. Vox Sang 1989;56:181–186.
for severely immunised mothers. Lancet 1979;i:604–605. 186. Haberman S, Blanton P, Martin J: Some observations on the
169. Bierme SJ, Blanc M, Fournie A, Abbal M: Desensitization by ABO antigen sites of the erythrocyte membranes of adults and
oral antigen. In Frigoletto FD Jr, Jewett SF, Konugres AA (eds): newborn infants. J Immunol 1967;98:150–160.
Rh hemolytic disease: New strategy for eradication. Boston: 187. Voak D, Williams MA: An explanation of the failure of the
GK Hall and Co, 1982:247–264. direct antiglobulin test to detect erythrocyte sensitization in
170. Barnes RMR, Duguid JKM, Roberts FM, et al: Oral adminis- ABO haemolytic disease of the newborn and observations on
tration of erythrocyte membrane antigen does not suppress pinocytosis of IgG anti-A antibodies by infant (cord) red cells.
anti-Rh(D) antibody responses in humans. Clin Exp Immunol Br J Haematol 1971;20:9–23.
1987;67:220–226. 188. Romano EL, Hughes-Jones NC, Mollison PL: Direct antiglob-
171. Huntley CC, Lyerly AD, Littlejohn MP, et al: ABO hemolytic ulin reaction in ABO-haemolytic disease of the newborn. Br
disease in Puerto Rico and North Carolina. Pediatrics Med J 1973;1:524–526.
1976;57:875–883. 189. Romans DG, Tilley CA, Dorrington KJ: Monogamous biva-
172. Bucher KA, Patterson AM, Elston RC, et al: Racial difference lency of IgG antibodies. I. Deficiency of branched ABHI-
in incidence of ABO hemolytic disease. Am J Public Health active oligosaccharide chains on red cells of infants causes
1976;66:854–858. the weak antiglobulin reactions in hemolytic disease of the
173. Kirkman HN Jr: Further evidence for a racial difference in fre- newborn due to ABO incompatibility. J Immunol 1980;
quency of ABO hemolytic disease. J Pediatr 1977;90:717–721. 124:2807–2811.
174. Peevy KJ, Wiseman HJ: ABO hemolytic disease of the 190. Osborn LM, Lenarsky C, Oaks RC, et al: Phototherapy in full-
newborn: evaluation of management and identification of term infants with hemolytic disease secondary to ABO incom-
racial and antigenic factors. Pediatr 1978;61:475–478. patibility. Pediatr 1984;74:371–374.
C H A P T E R 1 4

Hemolytic Transfusion
Reactions

A hemolytic transfusion reac- any clinical data other than a statement that they have
tion (HTR) should be defined caused HTRs.
as decreased RBC survival fol- Hemolytic transfusion reactions can occur in two
lowing blood transfusion. It is forms: immediate and delayed. Immediate HTRs
usually the transfused donor (IHTRs) occur during or immediately after the trans-
RBCs that are destroyed, but fusion (i.e., within hours). Delayed HTRs (DHTRs)
on rare occasions, it can be the can occur days or weeks following the transfusion;
recipient’s RBCs. If this defini- they usually occur when patients have previously
tion is used, then the signs of been sensitized to the offending antigen but have no
an HTR could range from antibody in their plasma at the time of cross-matching
obvious signs such as hemoglobinuria/hemoglobine- or transfusion. On transfusing RBCs containing the
mia and jaundice to a poor post-transfusion increment offending antigen, a secondary response occurs. This
in hemoglobin and hematocrit. Minimal shortening of response can occur as early as 24–48 hours after the
RBC survival might not be clinically obvious and transfusion but usually takes 5–14 days before it is
shown only by studies such as 51Cr RBC survival tests. noticed. The antibody that is produced sensitizes the
An HTR should not be defined by a positive direct transfused RBCs having the offending antigen; the
antiglobulin test (DAT) alone. Patients can develop a patient develops a positive DAT, and the RBCs might
positive DAT without any other signs of HTR; or might not have a shortened survival associated
although it is strictly true that such patients have suf- with clinical, hematologic, and/or biochemical signs.
fered a reaction to the transfused RBCs (i.e., a transfu-
sion reaction) because the RBCs are now sensitized
with antibody and/or complement (a “serological” INCIDENCE OF ALLOIMMUNIZATION
transfusion reaction), the sensitized RBCs often can
survive normally. For this reason, a hemolytic transfu- Antibodies that appear to react (e.g., sensitizing RBCs
sion reaction cannot be said to have occurred. to react by the antiglobulin test) at 37ºC and that are,
Unfortunately, some reports in the literature do not therefore, of potential clinical significance, have been
allow one to judge which definition of HTR the reported to occur in 1%–3.5% of hospital patients.1-9
authors have used; this is especially true when tables Hoeltge and colleagues8 reported that 2.9% of
contain many alloantibodies listed together without 4700 patients had alloantibodies but only 2% were of

541
542 Immune Hemolytic Anemias

potential clinical significance. Heddle and coworkers9 It is difficult to calculate the true rate of alloimmu-
followed 2490 patients given 11,218 red cell transfu- nization of multitransfused patients (including
sions. They found that 2.6% made alloantibodies if patients with SCD), as most of the studies just men-
they had no antibodies before the transfusion, tioned calculated their incidence based on antibodies
whereas 3.5% had alloantibodies even before transfu- detected after transfusion (without relating to the
sion. If the recipients had at least one antibody before presence of these antibodies before transfusion) and
transfusion, then 8.9% made new antibodies. If did not give details of their antibody detection
selected populations (e.g., following massive or multi- methods. This is particularly relevant to SCD, where
ple transfusions) are examined, the incidence of Lewis antibodies are often present as naturally occur-
alloantibodies can be much higher. ring antibodies. Other problems are that only a few of
Alloimmunization has been reported to occur in the SCD reports segregated patients who had received
0%–34% (mean of 12.5%, median of 11%) of 3347 blood matched for certain antigens from those who
random multitransfused patients.10-16 These patients received random units. When available, comparison
were transfused for open heart surgery, gastrointesti- of such data is of great interest. For instance, Wayne
nal bleeds, renal disease, transplants, and hematologi- and coworkers35 found an alloimmunization incidence
cal conditions. Two reports found that no patients with of 17% among patients receiving random units but
chronic lymphocytic leukemia made antibodies.12,13 only 6.4% among those receiving units matched for
The highest rate of alloimmunization recorded (34%) Rh, K, MNSs, Fya, and Fyb. Tahhan and associates36
was by Lostumbo and associates,10 who looked for found an incidence of 35% among patients receiving
antibodies in a defined time frame (one sample in the random units but 0% among patients receiving units
second postoperative week and another sample 4–12 matched for C, E, K, S, Fya, and Fyb. Most of the
months after surgery) and happened to use RBCs that reports agree that increasing alloimmunization paral-
were Lu(a+) for their antibody screen. Their tests lels increasing transfusions. Reisner and colleagues29
included a 22ºC phase and a papain test, in addition to found that the incidence of HTRs was 10% after 50
an antiglobulin test. They made the unique observation transfusions, 46% after 100 transfusions, and 57%
that anti-Lua was the most common specificity encoun- when 100–199 transfusions were given. Almost all
tered; the anti-Lua appeared earlier than 5 weeks after patients who are going to make antibodies make them
transfusion and then disappeared. If one subtracts the by the 10th to the 15th transfusion.
number of anti-Lua and clinically insignificant anti- The alloimmunization rate among patients with
bodies in the Lostumbo and colleagues report,10 the AIHA is high. It has not been so obvious as the high
rate of alloimmunization would be 10%, which is rate reported for SCD, as in AIHA the presence of
similar to that described in other reports. alloantibodies is often masked by autoantibodies.
In six reports, 2714 thalassemias were found to have When adsorption of autoantibodies is carried out, the
an alloimmunization rate of 3.7%–23% (mean of 12%, alloimmunization rate has been reported as 15%–41%.
median of 7.7%).17-23 It has been suggested that the Wallhermfechtel and coworkers,38 Issitt and associ-
lower rate of immunization among patients with tha- ates,39 and Leger and Garratty40 performed adsorp-
lassemia compared with patients with sickle cell tions with allogeneic RBCs and found alloantibodies
disease (SCD) (see the discussion later in this chapter) in 15% (19/125), 41% ([14 of 34] and 44% [299 of 674])
is because patients with thalassemia start transfusions of sera, respectively. In studies using adsorptions with
at an earlier age than those with SCD. Michail- autologous RBCs, Issitt and associates,39 James and
Merianou and colleagues20 showed that thalassemic colleagues,41 Laine and Beattie,42 and Morel and
children who started transfusions at an early age had coworkers43 detected alloantibodies in 27% (11 of 41),
an alloimmunization rate of 7.7%, compared with a 32% (13 of 41), 38% (41 of 109), and 40% (8 of 20) of
rate of 28% among children who started transfusions sera, respectively. Sokol and colleagues44 detected
later in life. It has also been argued that, compared alloantibodies in 14% (294 of 2149) of their patients,
with SCD patients, the donor blood given to patients but that series included samples from patients with
with thalassemia is usually from a more genetically warm- and cold-reactive autoantibodies and did not
similar population.23,24 distinguish how many patients with warm autoanti-
The highest incidence of alloimmunization appears bodies had alloantibodies. Sokol and colleagues44 also
to be associated with SCD and AIHA. In 12 reports, stated that they issued K–, Rh phenotype-matched
alloimmunization occurred in 8%–35% (mean 25%, blood to decrease the incidence of alloimmunization,
median 25%) of 2818 transfused SCD patients.25-37 so this study cannot be compared directly with the
Anti-E (21%), -C (14%), and -K (14%) were, by far, the others. Issitt and colleagues39 found that 69% and
most common antibodies encountered. Anti-Fya (7%), 19% of the suspected alloantibodies detected after
-Jkb (5%), and -S (4%) were the next most common adsorptions with autologous and allogeneic RBCs,
antibodies of possible clinical significance. Lewis respectively, had autoantbodies that mimicked allo-
system antibodies were also common (11%), but it was antibodies (see Chapter 7).
unclear from the reports whether they were stimu- As with SCD, a major factor is the number of trans-
lated by the transfusions or, more probably, were fusions the patients have received previously. James
present before transfusion. and coworkers41 and Wallhermfechtel and associates38
Hemolytic Transfusion Reactions 543

found that 75% (3 of 4) and 32% (6 of 19), respectively, TABLE 14-1. POTENTIAL CLINICAL SIGNIFICANCE
of patients who had received more than five transfu- OF 37°C-REACTIVE ANTIBODIES
sions had alloantibodies present. Such a high rate of
alloimmunization in patients with warm autoantibod- HDN HTR 51Cr
ies underscores the need for efficient techniques to
evaluate all sera from patients with AIHA for alloanti- Rh + + +
bodies before transfusion (see Chapter 10). Kell + + +
Duffy + + +
ANTIBODIES HAVING POTENTIAL Kidd + + +
SsU + + +
CLINICAL SIGNIFICANCE
Lea 0 + +
Lua (+) 0 0
Alloantibodies are of potential clinical significance Lub (+) + +
only if they react at 37ºC. Antibodies that react, Wra + + +
however strongly, at temperatures below 37ºC (e.g., at Vel (+) + +
room temperature) but not at 37ºC (e.g., most anti-A1, PP1Pk (Tja) + + NA
-M, -N, -P1, -Lea, -Leb, -I, -HI) will not cause HTRs.1,45-51 Holley (Hy) + + NA
It should be emphasized that if antibodies of any of Yta 0 ± ±
the specificities mentioned in the previous sentence
Leb 0 (+) 0
react at 37ºC, then they must be treated as being
Sda 0 (+) (+)
potentially clinically significant; indeed, some of them
Xga 0 (+) 0
have caused HTRs (e.g., anti-M, -P1, -Lea), but only
Cha 0 0 0
when they reacted at 37ºC. When an antibody reacts at
Yka 0 0 0
37ºC, it might or might not be of clinical
significance.1,45,51 Most antibodies should be consid- HDN = Antibody has caused hemolytic disease of newborn.
ered to have potential significance, but some HTR = Antibody has caused hemolytic transfusion reaction.
51Cr = Decreased RBC survival study using RBCs labeled with
specificities have never been proven to be the cause of 51Cr.
an HTR (e.g., anti-Bg, -Chido, -Xga); sometimes, blood + = Good evidence indicating potential clinical significance.
that is incompatible in vitro at 37ºC has been shown to (+) = Limited data (e.g., single case report) suggestive of clinical
significance (eg, a case of mild HDN in which the mother had
survive normally in vivo.1,45,51 Antibodies of some anti-Lub in her serum, but ABO HDN was not excluded; one
specificities are known to cause no RBC destruction in case in which anti-Sd a destroyed RBCs, but this particular
donor had a “super” Sd a antigen).
vivo sometimes but on occasion have been shown to ± = Some examples have been shown to have clinical significance;
cause in vivo destruction (e.g., anti-Lub, -Yta, -Ytb, others have been shown to have no clinical significance.
-Ge, -Lan).44,45 Table 14-1 summarizes the potential 0 = Evidence (or lack of reports) in the literature suggests that the
antibody has no clinical significance.
clinical significance of some commonly (and less com- NA = Data not available.
monly) encountered blood group alloantibodies. The
table is a summary of a literature review on antibod-
ies that have been proven to cause hemolytic disease
of the newborn (see Chapter 13), HTRs, and in significant antibodies detected in 214,000 patients
vivo destruction of small amounts (e.g., 1 mL) of from two hospitals over a 16-year period.
51Cr-labeled RBCs. Increased destruction of these In the four series, which included 21,499 antibodies
small amounts of 51Cr-labeled RBCs does not always detected at 37ºC, in blood recipients from three differ-
mean that larger volumes (e.g., 500 mL) will have ent countries, the findings were as follows: Anti-D
decreased survival.1,51 was the most common antibody detected (in all four
Table 14-2 shows data extracted from four large series), followed by anti-C when together with anti-D,
series of 37ºC-reactive alloantibodies that were and anti-E (approximately the same incidence); then
detected in hospital patients. The data, reported by anti-c and anti-Fya, followed by anti-Jka, anti-e, and,
Grove-Rasmussen,3 were from 18 large transfusion finally, anti-S. These results from data gathered before
centers in the United States, where 1.5 million units of 1977 might not reflect the impact of Rh prophylaxis.
blood were transfused. Anti-D was the most common Studies published in the 1990s could reflect the
antibody detected; the next most common specificity current trend more accurately. In the United States,
was anti-E, followed by anti-K, anti-c, anti-Fya, Hoeltge and coworkers8 studied alloimmunization in
anti-Jka, and anti-e. Spielman and Seidl5 reported on 4700 patients during the period from 1985 to 1993.
antibodies detected in 55,350 blood recipients in They found the most common specificities were anti-
Germany; 443 antibodies were detected (0.8% of all K (23%), anti-E (18%), anti-D (12%), anti-Lea (7.3%),
recipients). Tovey4 reported on 3707 antibodies anti-C (6.3%), anti-Fya (5.7%), anti-c (4.4%), and anti-
detected over a 5-year period in England; 3002 were Jka (3%). Heddle and associates9 (Canada) found that
from Rh-negative individuals and contained anti-D the most common antibodies made by 2490 patients
with or without anti-C, anti-E, or anti-K. Walker were anti-Jka(16.4%), anti-E (16.4%), and anti-K
and colleagues6 reported on 1537 (0.7%) clinically (14.6%).
544 Immune Hemolytic Anemias

TABLE 14-2. 37°C-REACTIVE ANTIBODIES DETECTED IN BLOOD TRANSFUSION RECIPIENTS IN FOUR STUDIES
FROM THE 1970S*

Grove- Spielman Walker Percentage Order of


Rasmussen3 Tovey4 & Seidl5 et al.6 Total of Total Frequency

Anti-D 8772 3002 245 778 12,797 61 1


-C (together with D) 2156 28 52 163 2399 11.4 2
-E 1079 231 45 118 1473 7 3
-K (-k) 978 174 41 181 1374 6.5 4
-c 619 154 14 53 840 4 5
-Fy (-Fyb) 372 72 16 55 515 2.4 6
-Jka (-Jkb) 141 17 12 13 183 0.9 7
-S -s 0 9 0 0 9 0.04 8
Others (e.g., anti-Lea, 1177 0 72 184 1433 6.9
-Leb, -Wra, -M, -N, -P1,
and unidentified)

*See text for comparative rates for the 1990s.

Most Common Antibodies INCIDENCE OF HTRs


to Cause HTRs
Table 14-3 shows data from three large studies from The reported incidence of HTRs in the transfused
the 1970s.3,6,52 The data are based on 280 HTRs (not population varies considerably in the literature.
including ABO) occurring after almost two million There are three main reasons for this. First, compati-
transfusions (incidence of one per 7143 transfusions). bility techniques have improved. Second, as will be
The most common antibody to cause an HTR was discussed shortly, the incidence varies depending on
anti-K, followed closely by anti-E. The next most the criteria that were used to select patients for a
common antibodies were anti-Fya, anti-c, and anti-Jka. particular series of HTRs. The third reason is that
It should be pointed out that the Grove-Rasmussen the incidence is dependent on the efficiency of the
report3 was a summary of data from 18 different monitoring system. The Mayo Clinic52-57 published
medical centers. The Mayo Clinic’s 10-year study52-55 six often-referenced reports relating to their experi-
also found anti-K to be the most common cause of ences of HTRs, but it should be emphasized that the
HTR, but in contrast to the Grove-Rasmussen study,3 earlier reports included asymptomatic patients (e.g.,
the Mayo researchers found anti-Jka to be almost as patients who developed a positive DAT following
common a cause of HTR, and they found anti-Fya transfusion but no clinical symptoms) among those
equal to anti-E as the third most common offenders. defined as suffering with HTRs. In one of the reports,
When DHTRs were examined separately, anti-Jka and 13 of 37 patients (35%) described as having HTRs had
anti-E were found to be the cause of HTRs more often no clinical symptoms at all.54 As there was no proven
than anti-K. Anti-Jka and anti-E were followed by decreased RBC survival in these patients, we would
anti-K and anti-Fya, respectively, as causes of DHTR. prefer not to include them as HTRs. Nevertheless, the
Unfortunately, it is not clear from the publications reports from the Mayo Clinic still contain the most
how these authors defined a hemolytic transfusion substantial hospital experience on HTRs available in
reaction (see the discussion that follows). the literature. During a 16-year period at the Mayo

TABLE 14-3. ANTIBODIES CAUSING HTRS

Grove-Rasmussen (1973)3 Walker et al. (1977)6 Pineda et al. (1978)52 Total

Units transfused 1.5 × 106 160,000 268,000 2 ×106


Number of HTRs 192 41 47 280
K 38 4 10 52
-E 30 10 7 47
-Fya 25 3 7 35
-c 25 1 6 32
-D 23 4 4 31
-Jka 8 4 9 21
-C (together with -D) 12 0 0 12
-e 4 0 4 8
Other antibodies (e.g., anti-k, -Fyb, 27 21 5 45
-Kpa, -Jsa, -Lea, -P1, -M, -Cw)
Hemolytic Transfusion Reactions 545

Clinic (1964–1980), 171 cases of HTRs were detected DELAYED HEMOLYTIC TRANSFUSION
following 649,943 transfusions (one reaction for REACTIONS (DHTRs) VS. DELAYED
every 3800 transfusions); if one disregards the SEROLOGIC TRANSFUSION REACTIONS
asymptomatic cases, the incidence would be approx-
imately one in 5700 transfusions. There were almost (DSTRs)
twice as many reactions among females compared
with males, and an increased frequency of reactions As mentioned previously, a transfusion reaction can
among older patients and among those with condi- be classified as an HTR only if evidence of hemolysis
tions requiring large amounts of blood (e.g., acute (i.e., reduced red cell survival) is documented. Ness
medical, obstetrical, and surgical situations). A more and colleagues60 coined the term “delayed serological
recent interpretation of the Mayo Clinic data is dis- transfusion reaction” (DSTR) to encompass those
cussed by Vamvakas and colleagues56 and Pineda reactions in which alloimmunization occurred,
and coworkers.57 leading to a positive DAT following transfusion but
The first clear report of a DHTR was by Fudenberg without any evidence of hemolytic anemia. They
and Allen.58 Since then, many have been reported; reported 34 cases of DSTR, 70% of which were due to
usually these have been single reports associated anti-E and/or anti-Jka, over a 20-month period.
with a particular specificity, but some series of Retrospective review of the medical records found
DHTRs have been reported.52-57 At the Mayo Clinic, clinical evidence of hemolysis in only six of the 34
DHTRs were reported to occur once in every 3200 (18%) cases; all six were associated with anti-Jka. Thus,
transfusions over a 16-year period; there was a great the incidence of DSTR was one per 151 (0.66%) recipi-
deal of variation in incidence from period to period ents with post-transfusion samples available for
(e.g., 1:11,650 from 1964 to 1973; 1:4000 from 1974 to testing, whereas the incidence of DHTR was only one
1977; and 1:1500 from 1978 to 1980).55 Along with the in 854 (0.12%) patients tested. Fifteen of the 34 patients
increased incidence of DHTRs since 1974 came a were followed for up to 174 days after the reaction.
decrease in IHTRs (1:12,000 from 1964 to 1973; Twelve of the 15 still demonstrated a positive DAT at
1:21,000 from 1974 to 1977; and 1:17,000 from 1978 to 174 days after transfusion. Eluate studies indicated
1980); the overall incidence of IHTRs from 1964 to that the persistence of a positive DAT after DSTR or
1980 was 1:14,000. More recent data from the Mayo DHTR could involve several immunologic mecha-
Clinic, reviewing two periods (1980–1992 and nisms, including the development of post-transfusion
1993–1998) and breaking the data into true hemolytic autoantibodies. Other workers (including those from
transfusion reactions and serological transfusion the Mayo Clinic) have presented transfusion reaction
reactions (see the discussion later), found an inci- data contrasting DHTRs with DSTRs.9,56,57,60,61
dence of one DHTR per 5405 and 6715 transfusions, Table 14-4 shows the results of five studies. It should
respectively.56,57 Taswell and colleagues55 point out be noted that Heddle and colleagues9 used a different
that the apparent increase in the incidence of DHTRs definition for DSTR; they included any patient who
at the Mayo Clinic since 1974 was probably due to became alloimmunized after transfusion, without
the following factors: necessarily having a positive DAT. Pineda and
colleagues57 studied two periods (1980–1992 and
1. Increased sensitivity of laboratory methods 1993–1998). The incidence of DHTR/DSTR increased
2. Increased awareness of DHTRs from one in 1899 in the period 1980 through 1992
3. Recognition and inclusion of milder, or entirely to one in 1300 in the period 1993 through 1998
asymptomatic, cases (e.g., positive DAT but no clin- (p < 0.05). Similarly, DSTRs increased from one in 2990
ical symptoms) in the period 1980 through 1992 to one in 1612 in the
4. Improved error recognition and control period 1993 through 1998 (p < 0.05). The incidence of
5. Increased experience and improved organization DHTR showed a trend toward decrease, from one in
(e.g., intravenous service) 5405 in 1980 through 1992 to one in 6715 in 1993
through 1998. The incidence of Jka antibodies
These reasons also probably explain why there is a increased in 1993 through 1998, while the incidence of
higher incidence of HTRs (especially DHTRs) other alloantibodies remained stable. These changes
reported by the Mayo Clinic than from other institu- were most likely due to a combination of factors,
tions. For instance, Croucher and associates59 including a decrease in average length of stay and the
reported an incidence of only one DHTR for every adoption of the polyethylene glycol (PEG) antibody
22,000 transfusions performed at a Toronto-based detection system.57
hospital; the Mayo Clinic incidence is about three to It is obvious from the data that most immune trans-
four times higher.59 The difference almost certainly fusion reactions associated with a positive DAT do not
does not mean that it is safer to receive blood in lead to any obvious clinical or laboratory signs of
Toronto than at the Mayo Clinic, but it does empha- hemolysis. Table 14-5 shows DHTRs or DSTRs, associ-
size how much variation exists, even between well- ated with various antibodies, at the Mayo Clinic over
respected workers, in definitions and recognition of a a 19-year period.56,57 Of immune-related transfusion
DHTR. reactions, 65% were DSTR (i.e., positive DAT after
546 Immune Hemolytic Anemias

TABLE 14-4. DELAYED HEMOLYTIC TRANSFUSION REACTIONS (DHTRS) VS. DELAYED SEROLOGICAL
TRANSFUSION REACTIONS (DSTRS)

Ness et al.60 Pinkerton et al.61 Heddle et al.9 Vamvakas et al.56 Pineda et al.57

DHTR
per unit 1/9094 1/13,680 1/11,328 1/5405b 1/9244c
per patient 1/854 1/2537 1/2082 NA NA
DSTR
per unit 1/1605 1/3040 1/199a 1/2990b 1/2312c
per patient 1/151 1/563 1/37a NA NA

NA, data not available


a
Heddle et al. included any patients who made alloantibodies following transfusion, even if the DAT was negative; other investigators used
positive DAT, following alloimmunization, to indicate DSTRs.
b Mayo Clinic data from 1980–1992.
c
Mayo Clinic data from 1993–1998.

transfusion but no signs of hemolysis), and 35% were Ness and coworkers60 reported that all 34 patients
true HTRs. This does not mean that the transfused in their study had a positive DAT. In contrast to
RBCs in the DSTR group survived normally; none of Salama and Mueller-Eckhardt,62 Ness and coworkers60
the studies included RBC survival studies. detected RBC-bound IgG by the DAT in all patients;
Nevertheless, the clinical effects were minimal in most 56% also had RBC-bound complement. Alloantibody
cases. The results of these studies also emphasize that was eluted from the RBCs of all patients. Fifteen of the
the term HTR has been applied incorrectly in the past 34 patients were followed for up to 194 days after the
and might still be so in some modern reports; a similar
situation applies to the term hemolytic disease of the
newborn.
TABLE 14-5. DELAYED HEMOLYTIC (DHTR)
In 1984, Salama and Mueller-Eckhardt62 published
AND SEROLOGICAL (DSTR) TRANSFUSION REACTIONS
an iconoclastic paper suggesting that some of our
AT MAYO CLINIC DURING A 19-YEAR PERIOD
cherished beliefs concerning the findings associated
(1980–1998)
with DHTRs were incorrect. Some people found these
suggestions hard to accept, but in 1990, Ness and col-
Specificity Total Number DHTR DSTR*
leagues60 published a study from the United States
that confirmed much of what Salama and Mueller- E 184 47 137
Eckhardt had found. Jka 95 45 50
Salama and Mueller-Eckhardt62 studied 26 patients Fya 62 26 36
K 62 16 46
with DHTRs. All patients were found to have C3d c 54 18 36
detectable on their RBCs after the reaction. The RBC- Jkb 27 12 15
bound C3d, detectable by the DAT, was detected for Fyb 12 9 3
weeks and even months after the transfusion. RBC- C 22 8 14
bound IgG was detected by DAT in only 10 of 26 S 7 4 3
e 12 3 9
cases (39%), but a radioimmunoassay detected IgG CW 5 3 2
on the RBCs in 16 of 17 patients tested. IgG alloanti- Yta 2 1 1
bodies could be eluted from RBCs of 25 of the 26 A1 2 1 1
patients. Kpa 1 1 0
Lua 1 1 0
In 1990, Ness and coworkers60 published results Lub 1 1 0
agreeing with the more iconoclastic parts of the article D 1 1 0
by Salama and Mueller-Eckhardt.62 Thirty-four trans- M 2 0 2
fused patients were studied. All of these patients had Jsa 2 0 2
serologic evidence (e.g., alloantibodies in the serum V 2 0 2
G 1 0 1
and a positive DAT) of a DHTR, but only five (18%) of P1 1 0 1
the 34 patients had clinical evidence of hemolysis. Cob 1 0 1
Ness and coworkers60 suggested that the term DHTR Total 559 197 (35%) 362 (65%)
be reserved for reactions occurring in patients who
show well-defined evidence of hemolysis, whereas *Alloimmunization with post-transfusion-positive DAT but no obvious
hemolytic anemia or clinical signs of a HTR.
the other patients should be described as having From Vamvakas EC, Pineda AA, Reisra R, et al: The differentiation of
DSTR. The incidence of a DHTR was one in 854 delayed hemolytic and delayed serologic transfusion reactions: Incidence
patients, and the incidence of a DSTR was one in 151 and predictors of hemolysis. Transfusion 1995;35:26–32 and Pineda AA,
Vamvakas EC, Gorden LD, et al: Trends in the incidence of delayed
patients. Results of other studies since then are shown hemolytic and delayed serologic transfusion reactions. Transfusion 1999;
in Table 14-4 and Table 14-5. 39:1097–1103.
Hemolytic Transfusion Reactions 547

putative transfusion. Twelve of the 15 (80%) still had intravascular coagulation (7%), and new RBC
positive DATs at 194 days after transfusion; at that alloantibodies that were not detectable until 72
time, RBC-bound IgG was detectable on the RBCs of hours or longer after the DHTR (7%) or that were
13 of the 15 patients, IgG plus C3 was detected on the never detected (20%).37,70-75 Some patients with SCD
RBCs of five patients (33%), and one patient had com- and DHTRs develop a life-threatening or fatal
plement on the RBCs only. Putative alloantibodies anemia (11%) with some, or all, of the unusual
were eluted from the RBCs of five (33%) patients findings just described.37
25–168 days after the last transfusion. Panagglutinins
(autoantibodies) were eluted from the RBCs of four
patients (27%); only specific alloantibodies were The Sickle Cell Hemolytic
found in the sera of these four patients. A mixed-field Transfusion Reaction Syndrome
appearance in the DAT was detected in only six We, and others, have observed serious and life-
patients (17.7%). threatening signs and symptoms that have occurred
These two reports have changed our expectations of in some patients with SCD who have severe HTRs.
the serology associated with DHTRs.60,62 The differ- Petz and colleagues76 described five such patients, and
ences from what we previously believed to be typical they felt that there are distinctive features of these
findings associated with DHTRs, were as follows: hemolytic episodes that justify the designation of a
syndrome that they have termed sickle cell hemolytic
• A mixed-field appearance was not observed com- transfusion reaction syndrome. In addition to the usual
monly in the DAT results. laboratory manifestations of hemolysis, patients can
• A positive DAT was detected for more than develop symptoms characteristic of a sickle cell pain
100 days after transfusion in five of seven patients crisis, and life-threatening anemia can develop as a
(71%) who were followed up for up to 312 days result of the hemoglobin and hematocrit falling to
after transfusion. levels markedly lower than were present prior to
• The alloantibody(ies) causing the DHTR could be transfusion. Other investigators have reported cases
eluted from the DAT-positive RBCs more than with many similar features, but insufficient emphasis
100 days after transfusion. has been given to the constellation of findings that
justifies the identification of a distinct syndrome. It is
The authors emphasized that because so few trans- important that the manifestations of this syndrome be
fused RBCs would be expected to survive 100 days recognized, as misinterpretation of the findings could
after transfusion, the alloantibody and complement lead to inappropriate management.
must be present on autologous RBCs. It was sug-
gested that the syndrome was very similar to post-
transfusion purpura (PTP).62 Characteristics of Sickle Cell
HTR Syndrome
HTRs ASSOCIATED WITH SICKLE The characteristics of sickle cell HTR syndrome are
CELL DISEASE indicated in Table 14-6, and certain aspects deserve
emphasis.
The literature contains many reports with detailed
case histories of HTRs associated with SCD; the SYMPTOMS OF A SICKLE CELL PAIN CRISIS
incidence of HTRs in SCD is not so well docu- LEADING TO ERRONEOUS DIAGNOSIS
mented.31,63-81 DHTRs in transfused SCD patients
have been reported in four series to have an incidence Symptoms suggestive of a sickle cell pain crisis fre-
of 4%, 11%, 17%, and 22%.29,30,32,64 The incidence of quently develop or are intensified during the hemo-
DHTRs in SCD is at least ten times higher than lytic reaction. Indeed, Garratty37 found that “pain
reported for random transfused patients. crises” developed in 87% of reported cases of delayed
Most of the reports of DHTRs in SCD follow the HTRs in patients with SCD. Chaplin and Cassell80
typical pattern of a DHTR, with new alloantibodies pointed out the relationship of HTRs to development
(Rh system mainly anti-C and -E [39%], -Jkb [15%], of pain crises in a remarkably detailed case report.
-Fya [10%], and anti-S [7% each], -K, -Fyb, and -s [3% They described a patient with SCD whose multiple
each]) appearing after 7 to 10 days, obvious signs episodes of post-transfusion hemolysis were docu-
(laboratory and clinical) of hemolytic anemia, a pos- mented with 17 measurements of in vivo RBC sur-
itive DAT, and decreased survival of the transfused vival. A most striking feature of the patient’s course
red cells. Many of the reports contain findings that was the regular onset of typical painful sickle cell
are not typical of DHTR: pain crisis (87%), post- crises coincident with the rapid destruction of large
transfusion hemoglobin or hematocrit dropping numbers of donor RBCs.
below pretransfusion levels (83%), hemoglobinuria Patients who experience brisk hemolysis often
and/or hemoglobinemia (33%), negative DAT develop fever, chills, flank pain, abdominal pain, chest
(26%), pulmonary infiltrates (9%), disseminated pain, apprehension, dyspnea, headache, nausea,
548 Immune Hemolytic Anemias

cases of HTRs in SCD, the post-transfusion hemoglo-


TABLE 14-6. COMPONENTS OF THE SICKLE CELL
bin and hematocrit fell below their pretransfusion
HEMOLYTIC TRANSFUSION REACTION SYNDROME
levels. The case report visualized in Figure 14-1 (left
panel) illustrates this point.
• Manifestations of an acute or delayed hemolytic transfusion
reaction.
• Symptoms suggestive of a sickle cell pain crisis that develop or P A T I E N T 1 : A 28-year-old female with SCD was hos-
are intensified during the hemolytic reaction. pitalized with chest, knee, and back pain, diarrhea, and an
• Marked reticulocytopenia (a significant decrease in absolute ankle ulcer. Her blood type was group A, Rh positive. She
reticulocyte level compared with the patient’s usual value).
• Development of a more severe anemia after transfusion than
had a history of multiple previous transfusions, and on
was previously present. A rapid drop in hemoglobin and admission her serum contained anti-E, -C, -K, and -S red cell
hematocrit can occur when hemolysis of donor RBCs is alloantibodies. The DAT was negative, LDH was 518 U/L,
accompanied by suppressed erythropoiesis, as sickle cell RBCs and total and direct bilirubin levels were 1.3 mg/dL and
have an intrinsically short survival. In some patients, it is 0.0 mg/dL, respectively. On hospital day 2, her hematocrit
possible that hyperhemolysis of autologous RBCs (bystander
immune hemolysis) could play a role in causing the decrease in was 13.2%, and she was transfused with 3 units of RBC
hemoglobin and hematocrit, although more definitive over the next 5 days, after which her hematocrit rose to
documentation of this phenomenon is necessary. 24.7%. All transfused RBCs were negative for E, C, K, and
• Subsequent transfusions could further exacerbate the anemia, S antigens and were crossmatch compatible.
which could become life threatening or even fatal.
• Patients often have multiple RBC alloantibodies and might also
have autoantibodies, making it difficult or impossible to find Her pain symptoms intensified, and on the seventh hos-
compatible units of RBCs. In other patients, however, no pital day, overt signs of a delayed HTR developed, includ-
alloantibodies are demonstrable, or patients might have ing a drop in hematocrit and increases in total and direct
alloantibodies for which antigen-negative RBCs are readily bilirubin to 3.1 mg/dL and 0.2 mg/dL, respectively. On
obtainable.
• Serologic studies might not provide an explanation for the day 9, she received 3 additional units of RBCs, but gross
hemolytic transfusion reaction. Even RBCs phenotypically hemoglobinuria developed, and her hematocrit dropped
matched with multiple patient antigens might be hemolyzed. precipitously to 11% on day 12. Two new RBC alloanti-
bodies, anti-Fya and -Jkb, were identified, and the DAT
From Petz LD, Calhoun L, Shulman IA, et al: The sickle cell hemolytic
transfusion reaction syndrome. Transfusion 1997;37:382—392. was positive with polyspecific and anti-IgG antiserums.
During the next 3 days, the patient received an additional
9 units of blood, all of which were negative for E, C, K,
vomiting, hemoglobinemia, and hemoglobinuria. S, Fya, and Jkb antigens and were crossmatch compati-
Many of these same symptoms can occur as part of a ble. This resulted in a transient increase in hematocrit to
sickle cell “pain crisis.” As a result, the diagnosis of an 19%, but the LDH increased to 3580 U/L; the total and
HTR could go unrecognized, and appropriate man- direct bilirubin increased to 7.7 mg/dL and 0.6 mg/dL,
agement could be delayed. Indeed, several groups of respectively; reticulocytes reached a nadir of 4.5%; and
investigators have pointed out that many HTRs in the hematocrit dropped to 9.3% on day 17. Thus, after
sickle cell disease patients are misdiagnosed as typical transfusion of a total of 15 units of RBCs, her hematocrit
“vaso-occlusive” or “aplastic” sickle cell crises.64,65,70 had decreased from 13% to 9.3%. Prednisone, 60 mg
Pain symptoms might simply be part of an HTR, or daily, was begun on day 15, and 2 more units of RBCs
the HTR might cause symptoms indistinguishable were transfused on day 17. Subsequently, signs of hemo-
from a true pain crisis.64-66,70,80,81 In either case, the crit- lysis decreased, and the hematocrit progressively
ical point is that the diagnosis of an HTR could go increased. During the hemolytic episode, the patient expe-
unrecognized because of the tendency to attribute all rienced an increase in pain that began to resolve on
signs and symptoms in an acutely ill patient with about day 17. She was discharged on day 24 with a
sickle cell disease to a diagnosis of pain crisis. This is hematocrit of 22.4%.
a frequent error, and the delay in making a diagnosis Twenty-seven months later, the patient was admitted
of an HTR contributes significantly to morbidity and with symptoms of pain in her chest, knees, and back,
to the probability of mortalilty. In this regard, the case and a nonproductive cough and fever (see Fig. 14-1,
described by Reed and colleagues81 is most dramatic, right panel). The hematocrit was 16.1%; the DAT was
as the patient’s fever and worsening pain after trans- negative; total and direct bilirubin were at 2.2 mg/dL
fusions were attributed to vaso-occlusion, her hemo- and 0.3 mg/dL, respectively; LDH was 386 U/L; and
globin dropped to 1.4 g/dL, and she expired; the reticulocytes were 10.8%. She was transfused with two
diagnosis of HTR was made postmortem. units of RBCs that were crossmatch compatible and neg-
ative for all six antigens as previously. The hematocrit
THE DEVELOPMENT OF MORE SEVERE ANEMIA rose to 25%, and the patient was discharged the next
THAN WAS PRESENT PRIOR TO TRANSFUSION day. She was readmitted 6 days later with persistent
pain, however, at which time the hematocrit was 12.7%;
One of the most important findings in the sickle cell total and direct bilirubin were 1.4 mg/dL and 0.2
HTR syndrome is the development of more severe mg/dL, respectively; and LDH was 509 U/L. The next
anemia than was present prior to transfusion, as has day, the hematocrit was 10.8% and reticulocytes
been indicated in a number of reports.37,61,65,69,74,75 reached a nadir of 2.8%. Two additional units of RBCs
Indeed, Garratty37 indicated that in 83% of reported resulted in a hematocrit of 19%, but the patient devel-
Hemolytic Transfusion Reactions 549

38 34
36 Hemolysis 32
34 30
32 28
30 26
28 24
Hematocrit (%)

26 22

Reticulocytes (%)
24 20
22 18
20 16
18 14
16 12
14 10
12 8
10 6
8 4
6 2
1 2 3 2 4 3 2 2 2 1 1
4 0
1 2 3 4 5 6 7 8 9 1011 1213141516 17181920 21222324 252627 1 2 8 9 1011 1213141516 17181920 21222324 25
Days
FIGURE 14-1. Clinical course of Patient 1, indicating Hct, uncorrected reiculocyte counts (%), and episodes
of hemolysis. Numbers in circles indicate the number of RBCs that were transfused. (From
Petz LD, Calhoun L, Shulman IA, et al: The sickle cell hemolytic transfusion reaction syndrome. Transfusion
1997;37:382–392.)

oped dark urine and there was again a progressive fall compared with the patient’s usual value. Reticulo-
in hematocrit to 11.8% on day 16. At that time, anti-D cytosis is a critical mechanism by which patients with
was also identified, although it had not been evident pre- SCD partially compensate for their shortened red cell
viously; a review of previous records from another hospi- survival. If erythropoiesis is suppressed in patients
tal revealed that the patient was a D mosaic. Two with a very short RBC survival, a rapid increase in
additional units of compatible RBCs resulted in a tempo- the severity of the anemia will occur. Accordingly,
rary increase in hematocrit to 17.8%, followed by a pro- if a patient with SCD has a severe HTR in which
gressive fall to 10.5% over the next 3 days, during which transfused RBCs are hemolyzed rapidly and, in addi-
time the total and direct bilirubin peaked at 4.5 mg/dL tion, the patient’s reticulocyte level is significantly
and 0.8 mg/dL and the LDH peaked at 1011 U/L. On depressed, severe and life-threatening anemia could
day 22, hemoglobin electrophoresis revealed only develop.
hemoglobin S, indicating that all donor RBCs had been
hemolyzed. Transfusions were discontinued because of
SEROLOGIC FINDINGS
the repeated episodes of post-transfusion hemolysis. The
patient’s hematocrit gradually improved to 15%, and The sickle cell HTR syndrome most often occurs in
signs of hemolysis and symptoms of pain gradually patients who have multiple RBC alloantibodies, at
abated. The patient’s DAT remained negative throughout, times in association with autoantibodies. In some
although flow cytometry on day 23 indicated that the patients, a newly detected RBC alloantibody develops
patient’s RBCs were weakly coated with IgG. It should be after transfusion as is typical in a DHTR, whereas in
noted that during the first hemolytic episode, her reticulo- other patients, the serologic findings do not provide an
cytes dropped to a nadir of 4.5%, and during the second explanation for the hemolysis. This could be because
episode, to 2.8%. no new antibodies become apparent during the DHTR
or, in some instances, because no alloantibodies or
autoantibodies are demonstrable at any time.
RETICULOCYTOPENIA The classic publication by Chaplin and Cassel,80
cited earlier, described a patient with SCD in whom
Another frequent finding in the sickle cell HTR syn- rapid destruction of transfused RBCs occurred
drome, as illustrated in the foregoing case report, is repeatedly despite entirely compatible crossmatch
reticulocytopenia, which we have defined as a results by a wide variety of serologic methods. Overt
significant decrease in the absolute reticulocyte level hemolysis occurred even after the transfusion of
550 Immune Hemolytic Anemias

crossmatch–compatible RBCs from two siblings Chaplin and Zarkowsky82 reported on four patients
whose blood types were identical to the patient’s with SCD who developed severe hemolysis after
with respect to numerous RBC antigens. These transfusion. Before transfusion, all four patients had
authors were the first to point out the regular onset of two or more alloantibodies as well as a positive DAT
typical sickle pain “crisis” coincident with the rapid and a RBC autoantibody in the serum. The hemoglo-
destruction of large volumes of donor RBCs. bin dropped to levels of 2.8 g/dL to 3.8 g/dL in three
Diamond and coworkers64 and Cullis and associ- patients, and the authors pointed out that these low
ates73 also described patients who had clinical and values occurred during periods of “relative reticulo-
laboratory findings of a DHTR, although the DAT and cytopenia.” Indeed, the nadir of hemoglobin values
antibody screen remained negative. were associated with corrected reticulocyte counts
Although DHTRs in patients with SCD have been (calculated from the authors’ data) of 3.52%, 1.1%, and
reported in which serologic findings do not explain 0.36%. The authors demonstrated autoantibodies in
the hemolysis, one must not think of this phenome- all patients and attributed the accelerated hemolysis
non as unique to patients with SCD. Numerous cases to AIHA, which could have been precipitated by
of hemolysis of transfused RBCs have been described alloimmunization associated with transfusion (see
as occurring in the absence of serologically detectable Chapter 9). In spite of severe anemia, all patients
alloantibodies in patients without SCD. recovered from the hemolytic episodes, cortico-
steroids being the mainstay of therapy.
Additional Reports of Severe HTRs Cummins and colleagues70 reported two DHTRs in
in Patients with SCD patients with SCD. Both patients developed multiple
alloantibodies and an autoantibody, and the nadirs
Diamond and colleagues64 reported DHTRs in three of hemoglobin values during the reactions were
patients with SCD. The patients developed clinical 2.6 g/dL and 3.0 g/dL, respectively. Both patients
features of painful crises, and the authors pointed out developed pain symptoms while hemolyzing, and the
that HTR should be suspected “when patients have authors emphasized that HTRs in patients with SCD
recurrent or severe sickle crises after transfusion.” often are mistaken for vaso-occlusive crises.
Millner and coworkers65 described ten patients with Solanki and McCurdy63 reported on five patients
sickle cell anemia who became acutely ill after blood with SCD who had DHTRs. Although multiple RBC
transfusion. The acute illnesses were attributed to alloantibodies were ultimately detected in all patients,
DHTRs although, in two cases, the immunohema- the responsible antibodies were sometimes unde-
tologic data were incomplete. Most of the patients tectable for as long as 72 hours after the reaction. In
developed symptoms characteristic of pain crisis three patients, painful crises could have been precipi-
during the hemolysis, and several developed reticulo- tated by the DHTR.
cytopenia and “profound anemia,” with the hemoglo- Several additional pertinent reports have appeared
bin level falling below pretransfusion levels and to only in abstract form and therefore are difficult to
values as low as 3.5 g/dL. Two patients died. The evaluate with certainty. Sosler and coworkers83
authors emphasized that “many of these reactions go described two multiply transfused patients with SCD
unrecognized, being diagnosed as sickle cell crises.” who developed severe AIHA after transfusion of
Cullis and associates73 described one patient who RBCs lacking the multiple antigens to which they had
had anti-E, -C, and –Jsb, and who received an exchange developed alloantibodies. In both patients, the sero-
transfusion with RBCs negative for these antigens, logic strength of the autoantibody reactivity increased
which increased her hemoglobin from 7.2 g/dL to after transfusion. Hemoglobin levels fell to nadirs of
9.2 g/dL. At this time, 53% of the hemoglobin was 1.5 g/dL and 4.0 g/dL, respectively. Both patients
hemoglobin S and 46% hemoglobin A; that is, the recovered after prednisone therapy.
hemoglobin S concentration was about 4.9 g/dL. The Friedman and associates72 reported on three children
patient developed severe joint pains, a DHTR ensued, with SCD who developed HTRs, with post-transfusion
and 6 days later the hemoglobin was only 3.0 g/dL. hemoglobin levels falling below pretransfusion levels
The authors assumed that the severe anemia was “despite high reticulocyte counts.” (No reticulocyte
caused by hemolysis of the allogeneic RBCs and counts are provided, and it is not clear whether or not
“hyperhemolysis” of the patient’s RBCs. No follow-up the authors are referring to corrected reticulocyte
hemoglobin electrophoresis was performed and counts.) Serologic evaluations revealed multiple allo-
no reticulocyte counts were reported, however. antibodies in only one of the patients, and in no case
Suppression of erythropoiesis could have resulted in a did serologic findings entirely explain the observed
drop in the hemoglobin S-containing RBCs from hemolysis. One of the patients died, and severe anemia
4.9 g/dL to 3.0 g/dL in 6 days, and all of the allogeneic from rapid hemolysis contributed to his death. The
RBCs could have been hemolyzed. The case is notable authors emphasized that avoidance of further transfu-
in that the patient’s DAT remained negative through- sion in these patients is vital, as continued transfusion
out and no previously undetectable RBC alloantibod- could be lethal.
ies could be found on repeated serum samples over the King and coworkers75 described five patients with
next 8 weeks. SCD who experienced a DHTR 7 to 19 days after
Hemolytic Transfusion Reactions 551

transfusion. Each patient had received an exchange anti-IH antibody, which reacted with group O RBCs at
transfusion of 4 to 5 units of Hb S-negative RBCs in 30°C and 37°C in both saline and 30% albumin media
anticipation of elective surgery or for treatment of an but did not react in a saline medium with group B
acute painful crisis or acute chest syndrome. Three of cells at 37°C. She was transfused with group B cells,
the patients developed positive DATs after the had no adverse reaction, and her post-transfusion
exchange transfusions, but new RBC alloantibodies hematocrit was 17.2%.
were identified that were thought to be responsible for Win and associates79 reported two cases of severe,
the HTR. Two of the patients had received donor life-threatening HTRs in patients with SCD after the
blood matched with the recipient’s phenotype for transfusion of compatible RBC units. One patient was
major antigens in the Rh, Kell, Kidd, Duffy, and MNS a 33-year-old woman who was referred for antenatal
systems and developed no new allantibodies but nev- care for her fourth pregnancy. She had received
ertheless developed a DHTR. numerous transfusions in the past. A decision was
Syed and colleagues74 reported the details of made to treat her with a gradual exchange transfusion
DHTRs in four patients with SCD. Three of the starting at 26 weeks’ gestation. The pretransfusion
patients had been treated with exchange transfusions. hemoglobin was 7.6 g/dL, the DAT was negative,
Hemoglobin electrophoresis data obtained for one reticulocytes were 180 × 109/L (normal range, 10–100
patient during a DHTR showed disappearance of × 109/L), and there were no atypical RBC antibodies.
hemoglobin A, and the authors concluded that, in She was transfused with 2 RBC units, and 4 days later
spite of the presence of a warm autoantibody on the she was readmitted with generalized musculoskeletal
patient’s RBCs, the hemolysis after transfusion was pain and a 2-day history of passing dark urine. The
caused by alloantibodies. DAT remained negative, and no RBC alloantibodies
Reed and associates81 reported a 32-year-old were found. In spite of transfusion of an additional
woman with SCD associated with hemoglobin C, who unit of RBCs, her hemoglobin fell progressively to a
was admitted to the hospital with pain involving the nadir of 4.8 g/dL, and reticulocytes decreased to one
back, chest, and legs. She had been transfused fre- tenth their previous value at 18 × 109/L. She recovered
quently, was 18 weeks pregnant, and had multiple after treatment with methylprednisolone and IVIG.
RBC alloantibodies including anti-E, -S, -Jkb, and Fy3. Analysis of the patient’s urine revealed both hemo-
She was transfused with 1 unit of RBCs, but 1 week globin A and hemoglobin S, which the authors inter-
later she had worsening pain and her hemoglobin had preted to mean that both allogeneic and autologous
fallen from 7.7 g/dL to 6.8 g/dL. She was transfused RBC were being destroyed as part of the HTR. No
with 2 additional units of RBCs, and 4 days later her data were provided, however, to indicate that the
hemoglobin fell from 6.0 g/dL to 4.9 g/dL, at which urine did not contain hemoglobin S regularly as a
time she quickly became obtunded. Her serum was result of her usual hemolytic state, and the authors
reddish-brown, the hemoglobin decreased further to pointed out that there is little information on the
1.4 g/dL, and platelets fell from 131,000/μL to urinary excretion of free hemoglobin in SCD. Marked
31,000/μL. No reticulocyte counts were performed. reticulocytopenia also occurred in the authors’ other
Two group O Rh-negative units, which were not patient during an acute episode of hemolysis, and
crossmatched, were administered as an emergency, the bone marrow aspirate in both patients revealed
but the patient expired. As indicated previously, her erythroid hyperplasia which, in association with
pain was attributed to vaso-occlusion, and the diag- reticulocytopenia, indicates ineffective erythropoiesis.
nosis of a HTR was made postmortem. The authors speculated that the reticulocytopenia
Campbell and coworkers78 described an 18-year-old was due to peripheral consumption by hyperactive
woman with SCD who was 37.5 weeks pregnant. Her macrophages.
blood type was group B, Rh-positive, her admission Tolano and coworkers84 performed an 11-year retro-
hematocrit was 18%, and she was transfused with 1 spective chart review of patients in the pediatric age
unit of group B, Rh-positive RBCs. After delivery of a group with discharge diagnoses of SCD and trans-
male infant, her clinical course was complicated by fusion reaction. They encountered seven patients
infections, for which she was treated with antibiotics (ages 6 to 17 years) who developed nine characteristic
and then discharged. Four days later, however, she episodes of sickle cell HTRs. All patients presented
presented with intense right femur pain and a hemat- with fever and hemoglobinuria. Symptoms of pain (in
ocrit of 16%. The next day the hemoglobin was the back, abdomen, and/or legs), which initially were
4.2 g/dL, and she was admitted for emergent transfu- ascribed to vaso-occlusive crisis, were present in all
sion and pain management. A decision was made to but one episode. The hemoglobin decreased from pre-
transfuse phenotypically matched RBCs. No exten- transfusion levels in eight of nine episodes, the nadir
sively phenotyped group B RBCs were available; ranging from 4.0 g/dL to 5.9 g/dL, with a median
instead, group O cells that matched the patient’s phe- value of 4.5 g/dL. Of note was the fact that the cor-
notype were transfused. After receiving approx- rected reticulocyte count at the time of the hemo-
imately 100 mL, however, she experienced chills and globin nadir ranged from 0.2% to 5.6%, with a median
an increase in temperature from 36.4°C to 39°C. value of 1.16%. The DAT was positive in only two
Serologic studies revealed that the patient had an IgM of the nine events at presentation. One patient had
552 Immune Hemolytic Anemias

previous alloantibodies, and four patients developed transplant conditioning, her hemolysis abated and her
new identifiable antibodies, which included a warm DAT became negative.
autoantibody in one patient. The new alloantibodies The findings of Sirchia and colleagues85 and
appeared at the time of the event in two patients but Grainger and associates86 are of considerable interest
were not detected for one month in a third patient. because they indicate that the mechanism of the
Severe complications included acute chest syndrome, severe hemolysis which can occur during HTRs in
pancreatitis, congestive heart failure, and acute renal patients with SCD is not related to the presence of
failure. Three patients received additional RBC hemoglobin S or SCD. Because the finding of a lower
transfusions that were “completely” phenotypically hemoglobin level after transfusion than that prior to
matched, one patient experienced severe additional transfusion has been noted after HTRs in patients
hemolysis. The patients responded to treatment with with two forms of hereditary hemolytic anemia, one
corticosteroids and erythropoietin. For reasons that wonders whether a similar occurrence does not some-
are not evident, the authors chose to exclude from times follow HTRs in patients with acquired
consideration patients who had acute HTRs, although hemolytic anemias.87
the syndrome can occur with either acute or delayed
HTRs (see Table 14-6). The Mechanism Underlying
Post-transfusion Fall in Hemoglobin
Similar Hemolytic Transfusion Reaction to Values Lower Than Pretransfusion
Syndromes in Patients with Thalassemia Values
If, following a transfusion, all of the transfused RBCs
Sirchia and colleagues85 reported their experience
were hemolzyed, one would expect the hemoglobin to
regarding HTRs that occurred in seven patients with
return to the pretransfusion level. In patients who
β-thalassemia. Their data indicate that findings
develop the sickle cell HTR syndrome, however, the
similar to those described for the sickle cell hemolytic
hemoglobin value after transfusion is significantly
transfusion reaction syndrome might occur in patients
lower than that prior to transfusion. Several possible
with other hemoglobinopathies. Four of the seven
mechanisms, which could be operating concomitantly,
patients who developed a HTR had a post-transfusion
have been proposed to explain this remarkable
hemoglobin level that was lower than the pretransfu-
finding.75,76,88
sion value (7.0–3.5; 5.0–4.0; 7.0–5.5; 4.6–3.0), but the
authors did not attempt to determine the mechanism
of this finding. HTRs occurred in spite of transfusion “HYPERHEMOLYSIS”
of RBCs that were compatible as indicated by exten-
sive compatibility test procedures. The serologic An increased rate of destruction of RBCs in a patient
findings after transfusion did not clearly explain the with a chronic hemolytic anemia can be termed hyper-
cause of the destruction of the transfused RBCs. High- hemolysis. (Unfortunately, this term is often used
dose steroids were administered to each of the seven simply as a synonym for severe hemolysis, rather than
patients, and they eventually showed a progressive referring to an increased, or hyper-, hemolytic rate.) If
resolution of the severe anemia manifested during the an HTR were to cause destruction of all of the trans-
HTRs. fused RBCs and also result in destruction of the
Grainger and associates86 reported a 1-year-old girl patient’s own RBCs, it is evident that the hemoglobin
with newly diagnosed β-thalassemia who developed would fall to a level lower than the pretransfusion
life-threatening hemolysis after transfusion. The value. In patients with SCD, the analysis is compli-
hemoglobin before transfusion was 5.3 g/dL, but 4 cated by the fact that the patient’s own RBCs inher-
weeks after an initial transfusion with red cells ently have a markedly short life span. Nevertheless, a
matched for Rh and Kell antigens, the hemoglobin had number of authors have suggested that hyperhemoly-
fallen to 3.5 g/dL and she had signs of hemolysis, sis is frequently the cause of or at least a contributing
including gross hemoglobinuria. Serologic findings factor in the development of the marked post-
typical of a delayed HTR were not present, although a transfusion fall in hematocrit observed in patients
weak cold autoantibody reactive up to 18°C was with SCD who have an HTR.37,76,81
found, and the DAT became positive with C3d on the Destruction of the patient’s own RBCs during an
cells. The hemolysis persisted, and her anemia pro- HTR could occur as a result of “bystander immune
gressed with subsequent transfusions, even though hemolysis”—immune lysis of RBCs by antibody
she was treated with methylprednisilone, IVIG, and directed against an antigen(s) that is not an intrinsic
splenectomy. One week after splenectomy, her hemo- component of the RBC membrane.86,87 In other
globin reached a nadir of only 1.9 g/dL. An HLA- words, the hemolysis of transfused RBCs by the
identical sibling donor was available, and the patient patient’s alloantibody results in autologous RBCs
was prepared for hematopoietic cell transplantation being hemolyzed as well. This topic is reviewed in
with cyclophosphamide and busulphan over the detail in the section on bystander immune cytolysis
course of 4 days. During the immunosuppressive in Chapter 9.
Hemolytic Transfusion Reactions 553

Development of AIHA in Patients Reticulocyte volume


with SCD = RBC volume × reticulocyte percentage
Another mechanism by which hemolysis of the
patient’s own RBCs could be destroyed during an Steady state daily RBC production/+4 destruction
HTR is the development of an autoantibody as a result Reticulocyte volume
of the transfusion, as reviewed previously in this =
Life span of reticulocytes (days)
chapter and in Chapter 9. Indeed, a number of authors
have suggested that autoantibodies that developed in
association with HTRs in patients with SCD were the (Blood volume) (hematocrit)
cause of hemolysis of the patient’s own RBCs.70,82,83,89 (reticulocyte percentage)
In other reported cases, however, autoantibodies were OR =
clearly not present, and for this reason they do not Life span of reticulocytes (days)
appear to be responsible for the post-transfusion fall
in hemoglobin in all cases.64,65,73,76 The maturation time of reticulocytes in the peripheral
blood is prolonged above the normal of approximately
1 day due to premature delivery of reticulocytes to the
SUPPRESSION OF ERYTHROPOIESIS circulation in patients with anemias who have marrow
stimulation. These reticulocytes, called “shift cells,”
In persons whose RBCs have a normal life span, the contain more reticulum than those in the normal subject
drop in hemoglobin after an HTR to a value signi- and require a longer period of time than normal circu-
ficantly lower than the starting level strongly suggests lating reticulocytes to lose their reticulum. Thus, the
the destruction of the patient’s own RBCs in addition index of daily RBC production/destruction must be
to the transfused RBCs. In patients with SCD or trans- “corrected” for the life span of reticulocytes in patients
fusion-dependent thalassemia, however, the patient’s with varying degrees of anemia by dividing the reticu-
own RBCs have a markedly short survival time, thus locyte volume by the life span of reticulocytes (in days).
complicating the analysis. Indeed, when patients Based on the data of Hillman and Finch,92 the following
with SCD develop suppression of erythropoiesis, correction factors can be applied: hematocrit 40 to 45,
such as can be caused by infection, a marked drop in correction 1.0; hematocrit 35 to 40, correction 1.5; hema-
hemoglobin is to be expected because of the short tocrit 25 to 35, correction 2.0; hematocrit lower than 25,
survival of the patients’ RBCs. Such an abrupt drop correction 2.5. Such corrections do not apply if erythro-
in hemoglobin and hematocrit can be mistaken for poiesis is suppressed, as it is, for example, by infection,
hyperhemolysis—a further shortening of the already in which reticulocyte life span is variable.
shortened life span of sickle cells. For example, if steady-state conditions for a 65-kg
Increasing the hematocrit by transfusion also female include a hematocrit of 22.5% and reticulocytes
causes suppression of erythropoiesis.90,91 Accordingly, of 20%, daily RBC production/senescence =
when there is an unexpectedly low hematocrit after
an HTR, it is important to calculate the magnitude of (65 kg) (64 mL/kg) (.225) (.20)
= 75 mL/day
the fall in hematocrit that can be explained on the 2.5 days
basis of suppression of erythropoiesis so as to not
interpret inappropriately the drop in hematocrit as
an indication of hyperhemolysis. The magnitude of
a fall in hematocrit that can be explained on the EFFECT OF SUPPRESSION OF ERYTHROPOIESIS
basis of depressed erythropoiesis without hyperhe- IN A PATIENT WITH SICKLE CELL ANEMIA
molysis can be calculated by taking into account the
expected loss of RBC volume by senescence while Petz and associates76 calculated the amount by which
also considering residual RBC production as indi- the hematocrit could have fallen on the basis of sup-
cated by hematologic data during this period. We pression of erythropoiesis in a patient with sickle cell
performed these calculations on several patients disease (Fig. 14-2). The volume of RBCs lost by senes-
who developed the sickle cell hemolytic transfusion cence was calculated using data on admission, as
reaction syndrome.76 these figures provide the best estimate of steady-state
conditions. The results of these calculations were con-
sistent with data reported by others for patients with
CALCULATION OF RBC PRODUCTION AND sickle cell anemia in steady-state conditions.93 In the
DESTRUCTION THROUGH SENESCENCE steady state, RBC production and destruction through
senescence are equal.
Blood volume in mL = Weight in kg × 69 mL/kg
(males) or 64 mL/kg P A T I E N T 2 : The patient was a 22-year-old male
(females) weighing 66 kg, who presented with a hematocrit of 25.9%
and a reticulocyte count of 18.1%. Calculation of daily RBC
RBC volume = Blood volume (mL) × hematocrit production and senescence using these data are as follows:
554 Immune Hemolytic Anemias

38 34
36 Hemolysis 32
34 30
32 28
30 26
28 24
Hematocrit (%)

26 22

Reticulocytes (%)
24 20
22 18
20 16
18 14
16 12
14 10
12 8
10 6
8 4
6 2
2 2 2 3 1 2 2 1 1 1 1
4 0
1 2 3 4 5 6 7 8 9 10 11 1213 1415 16 1718 1920 212223 24 2526 2728 29 3031 3233 34 3536 3738 39 40 41 52 62
Days
FIGURE 14-2. Clinical course of Patient 2, indicating Hct, uncorrected reticulocyte counts (%), and
episodes of hemolysis. Numbers in circles indicates the number of unites of RBCs that were transfused.
(From Petz LD, Calhoun L, Shulman IA, et al: The sickle cell hemolytic transfusion reaction syndrome.
Transfusion 1997;37:382–392.)

=Weight 66 kg period, the predicted RBC volume and hematocrit on day


Period of time
= 7 days 8 would be:
Initial hematocrit
= 25.9 %
Initial reticulocytes
= 18.1% RBC volume = 1179 mL – 749 mL = 430 mL
Initial blood volume
= (66 kg ) (69 mL/kg)
= 4554 mL 430
Hematocrit = = 9.4%
Initial red cell volume = (4554 mL) (.259) = 1179 mL 4554

Daily RBC production/senescence The measured hematocrit on day 8, however, was


(4554 mL) (.259) (.181) 13.9%, indicating an RBC volume on that day of 633 mL
= = 107 mL/day
2.0 days (4554 mL × 0.139), which is 203 mL higher than pre-
dicted on the basis of zero erythropoiesis. This indicates
By day 8, the hematocrit had fallen to 13.9%. During an average RBC production of 29 mL/day (rather than
this time, the patient had an infection manifested by 0 mL/day).
fever, productive cough, purulent sputum, and dyspnea. Production of 29 mL/day is consistent with calculated
Suppression of erythropoiesis, presumably a result of RBC production during this period of time. A hematocrit
infection, was indicated by a drop in the uncorrected and reticulocyte count are available on day 7 and,
reticulocyte count to 3.2% by day 7. The following are using these values, calculated RBC production would be
the pertinent calculations concerning the mechanism of 31 mL/day if the life span of reticulocytes was 1 day.
the patient’s marked drop in hematocrit.
For the 7-day period, the volume of RBCs lost through RBC volume day 7 = (4554 mL) (0.213) = 970 mL
senescence equaled
(970) (0.032)
RBC production = = 31 mL
(107 mL/day) (7 days) = 749 mL 1 day

Therefore, if RBC production dropped to zero and the If the life span of the patient’s reticulocytes was
RBC life span was unchanged throughout this 7-day 1.5 days, 2.0 days, or 2.5 days, the corresponding
Hemolytic Transfusion Reactions 555

values for RBC production/day would be 20.6 mL, 15.5 who develop an HTR is provided by the fact that a
mL, and 12.4 mL, respectively. similar fall in hemoglobin has been reported during
These calculations indicate that if the patient had been HTRs in patients with thalassemia.85 Suppression of
transfused on day 1, and if all allogeneic RBCs had been erythropoiesis or development of autoantibodies could
hemolyzed in an HTR but the life span of autologous RBCs occur in either SCD or thalassemia but, of course, sickle
had remained unchanged (that is, if there was no hyper- cell crisis is not common to both entities.
hemolysis), the hematocrit could have fallen alarmingly
from 25.9% to 13.9% over a 7-day period simply due to
suppression of erythropoiesis. Management of Patients with Sickle
Cell HTR Syndrome
One may conclude from these calculations that
when a patient with SCD develops reticulocytopenia The optimal management of patients with this syn-
during an HTR, suppression of erythropoiesis is likely drome is not yet defined.98 Because continued trans-
to be a significant contributing factor in the develop- fusion could lead to a further fall in the hemoglobin
ment of the marked post-transfusion fall in hematocrit and hematocrit, it is appropriate to discontinue trans-
and hemoglobin. fusion unless the hemoglobin is so low as to be life
threatening. Corticosteroids appear to be helpful
when given in doses of 1–2 mg/kg/day of prednisone
or equivalent.76 In addition, IVIG and erythropoietin
Sickle Cell Crisis as an Explanation may be beneficial.98
for the Post-transfusion Fall in Sirchia and colleagues85 administered high-dose
Hemoglobin during an HTR steroids to each of the seven patients with thalassemia
Some authors suggest that hyperhemolysis is a cardi- that they reported, and all patients showed a progres-
nal manifestation of painful sickle cell episodes.94 sive resolution of the severe anemia that developed
Hematologic studies on sickle cell patients during during HTRs. They cautioned, however, that addi-
pain crises do not support such a conclusion, tional factors could have contributed to the positive
however. Diggs95 performed daily reticulocyte counts outcome in their patients, so that the value of steroid
and hemoglobin determinations during 747 episodes therapy was uncertain.
of sickle cell pain crisis in 166 subjects. He also mea-
sured serum bilirubin and determined the output
of fecal urobilinogen. He concluded that a true PATHOPHYSIOLOGY OF HTRs
hemolytic crisis was not observed in any patient with
SCD. He observed that in the majority of patients with Most HTRs are associated with destruction of extravas-
SCD and secondary viral, bacterial, or protozoal infec- cular RBCs. It is rare for any of the antibodies listed in
tions and with systemic diseases, there is a decrease in Table 14-3 to activate complement efficiently enough to
RBC values, and this is primarily the result of defec- cause complement-mediated intravascular lysis, but on
tive regeneration rather than of an increase in the occasion it has occurred, especially with examples of
hemolytic rate. He concluded, rather emphatically, Kidd antibodies. Complement-mediated intravascular
“The concept of ‘hemolytic crisis’ in SCD is a myth hemolysis is usually associated with HTRs caused by
that should not be perpetuated in the light of present ABO antibodies (anti-A, -B, -H). Although Rh anti-
knowledge.” In a subsequent report, he restated: “The bodies, however strong, do not activate complement,
use of the term ‘hemolytic crisis’ as a synonym for hemoglobinemia and hemoglobinuria are sometimes
recurrent painful and febrile crises also is not justified. found in Rh-mediated HTRs. This is not a sign of com-
There is usually no significant decrease in erythro- plement-mediated intravascular lysis, but rather a con-
cytes at the time of painful crises compared with sequence of massive extravascular destruction. It could
asymptomatic intervals.”96 be that such destruction leads to release of hemoglobin
Further data were published by Naumann and into the peripheral circulation because of fragmenta-
coworkers,97 who measured plasma hemoglobin tion of RBCs during phagocytosis, and possible
during painful crises in patients with SCD. Although macrophage-induced cytotoxicity (see Chapter 4). On
they concluded that hyperhemolysis did occur during rare occasions, hemoglobinuria and hemoglobinuria
the first 2 days of crisis, the amounts of plasma hemo- are due directly to blood group antibodies (e.g., Kidd
globin detected could be accounted for by the destruc- antibodies). Beauregard and Blajchman99 have
tion of small volumes of RBCs. Indeed, the highest reviewed conditions that can mimic HTRs.
plasma hemoglobin value observed could have been To understand what happens in vivo when comple-
produced by lysis of only 20 mL of blood, which obvi- ment-mediated intravascular lysis occurs (e.g., by
ously would have no measurable effect on hemoglo- ABO antibodies), one has only to understand the clas-
bin or hematocrit determinations. sical pathway of complement activation described in
Compelling evidence that sickle cell crisis per se is Chapter 4. Most other antibodies (e.g., Rh, Kidd, Kell)
not an adequate explanation for the remarkable drop in destroy RBCs by extravascular mechanisms as des-
hemoglobin and hematocrit that occur in SCD patients cribed in Chapter 4.
556 Immune Hemolytic Anemias

Table 14-7 lists the symptoms and Table 14-8 the lab- which act on smooth muscle and interact with certain
oratory findings most commonly associated with HTRs. cells (e.g., mast cells) to release histamine. These effects
Figure 14-3 shows the temporal relationship of the lab- play a major role in symptoms such as flushing of the
oratory test results to the HTR, and Table 14-9 indicates skin, difficulty in breathing, and chest pain, which often
the volume of lysed RBCs required to produce grossly are associated with HTRs due to complement-activating
pink-red plasma. It should be noted that some of the antibodies. The most common abnormal laboratory
symptoms do not occur if the patient is anesthetized. In findings were decreased serum haptoglobin (92% of
the Mayo Clinic series,52-55 the most common symptoms cases); positive (usually weakly or transiently positive)
associated with DHTRs were fever and chills (75% and DATs (89% of cases; all negative DATs were associated
34%, respectively). Anemia (usually ≥2 g/dL below pre- with ABO immediate HTRs); and elevated serum biliru-
vious level), which was seen in 87% of DHTRs, was the bin (80% of cases).52-55 Figure 14-3 shows the temporal
leading complication.53 Oliguria and renal failure were relationship of some of these test results.100
noted in 34% and 13% of patients, respectively.52-55 If the
antibody causing the reaction is capable of activating
complement (e.g., ABO), the clinical symptoms could be Interrelationships of Mediators
different from those associated with noncomplement- of the Inflammatory Response
activating antibody (e.g., Rh). During complement acti-
vation, anaphylotoxins (C3a and C5a) are released, We have known for some years that some of the clini-
cal signs associated with HTRs are associated with
activation of the complement system, (e.g., produc-
tion of anaphylotoxins [C3a and C5a] and interaction
TABLE 14-7. SYMPTOMS OF DELAYED HEMOLYTIC between the complement and coagulation systems).
TRANSFUSION REACTIONS Until recently, it has been a puzzle why similar
symptoms could occur with HTRs associated with
Percentage of Patients Showing antibodies that do not activate complement. A
Symptom These Symptoms
number of studies have linked some of these symp-
Fever (≥2°F increase) 75 toms to a class of biologic mediators known as
Chills 34 cytokines.101-105 Cytokines are hormone-like glycopro-
Chest pain 13 teins secreted by a wide variety of cells, including
Hypotension 13
Nausea 4
lymphocytes, monocytes, macrophages, fibroblasts,
Flushing 4 and endothelial cells. Davenport and cowork-
Dyspnea 4 ers101,102,104,105 showed that several cytokines were
Hemoglobinuria 2 produced when ABO- or Rh-incompatible RBCs were
From Pineda AA, Brzica SM, Taswell HF: Hemolytic transfusion reaction.
added to anti-A/B or anti-D, respectively, in vitro.
Mayo Clin Proc 1978;53:378; Pineda AA, Taswell HF, Brzica SM: These cytokines included some interleukins (IL-1,
Delayed hemolytic transfusion reaction. Transfusion 1978;18:1; Moore SB, IL-6, IL-8), an interleukin receptor antagonist (IL1ra or
Taswell HF, Pineda AA, et al: Delayed hemolytic transfusion reactions. A M J
Clin Pathol 1980;74:94; and Taswell HF, Pineda AA, Moore SB: Hemolytic IRAP), tumor necrosis factor-alpha (TNF-α), and
transfusion reactions: Frequency and clinical and laboratory aspects. In Bell monocyte chemoattractant protein (MCP-1).
CA (ed): A Seminar on Immune-mediated Cell Destruction. Washington, DC: When ABO incompatibility was produced in vitro,
American Association of Blood Banks, 1981:71.
Davenport and coworkers101,102 found that IL-8 was
first observed after 2 hours and increased over a
24-hour period. Plasma TNF levels were maximal at
TABLE 14-8. LABORATORY ABNORMALITIES 2 hours and declined to control levels by 24 hours.
FOLLOWING HEMOLYTIC TRANSFUSION REACTIONS MCP-1 was detected at 24 hours but not at 6 hours.
Complement was necessary for optimal production of
Percentage of Patients Showing IL-8 and TNF but was not necessary for MCP-1. Some
These Abnormalities of the findings have been supported by an in vivo
event; Butler and associates103 studied a patient who
Low haptoglobin 92 was group O and accidentally received a group A unit;
Positive DAT 89a
Increased indirect bilirubin 80 TNF levels were found to increase 14-fold after the
Hemoglobin detected in 70b incompatible transfusion.
plasma and/or urine When IgG (anti-D)-mediated in vitro incompatibil-
a
All negative DATs were associated with ABO immediate HTRs.
ity was studied, Davenport and colleagues104,105
b 88% of immediate HTRs and 52% of DHTRs. found that IL-1, IL-6, IL-8, and TNF were produced.
From Pineda AA, Brzica SM, Taswell HF: Hemolytic transfusion reaction. The interleukins were detectable in the plasma at 4 to
Mayo Clin Proc 1978;53:378; Pineda AA, Taswell HF, Brzica SM:
Delayed hemolytic transfusion reaction. Transfusion 1978;18:1; Moore SB,
6 hours and increased up to 24 hours. TNF production
Taswell HF, Pineda AA, et al: Delayed hemolytic transfusion reactions. A M J peaked at 6 hours. MCP-1 was detectable at 24 hours
Clin Pathol 1980;74:94; and Taswell HF, Pineda AA, Moore SB: Hemolytic but not at 6 hours. An interleukin-1 receptor antago-
transfusion reactions: Frequency and clinical and laboratory aspects. In Bell
CA (ed): A Seminar on Immune-mediated Cell Destruction. Washington, DC: nist (IL-1ra) was also produced after IgG-mediated
American Association of Blood Banks, 1981:71. RBC incompatibility. IL-1ra production was evident at
Hemolytic Transfusion Reactions 557

Time after transfusion reaction


Test Immediate 1–3 hr 6–12 hr 24 hr Days

Blood

Hemoglobinemia

Agglutinates

DAT
FIGURE 14-3. Temporal results of
biochemical and serological tests following a Bilirubin
hemolytic transfusion reaction. (From
Crookston JH: Transfusion reactions. Am J Methemalbumin
Med Technol 1968;334:579–588.)
Haptoglobin

Urine

Hemoglobin

Urobilin

Hemosiderin

TABLE 14-9. VOLUME OF LYSED RBCs REQUIRED be aware that when a patient is undergoing surgery
TO PRODUCE GROSSLY PINK-RED PLASMA and being transfused, abnormal bleeding and/or
hypotension might be the only observable sign of an
Hb Content
HTR due to ABO-incompatible blood. The exact rela-
tionship between HTRs and DIC is uncertain.
Product g mg Rare cases of DIC have been described following
noncomplement-mediated extravascular RBC destruc-
100 mL whole blood 15 15,000 tion; recent data suggest that these could be due to
100 mL packed RBCs 33 33,000
1 mL packed RBCs 0.33 330
release of cytokines, which, like complement, can
5 mL packed RBCs 1.65 1650 interact with the coagulation pathway. Capon and
Goldfinger106 believe that cytokines could play a major
Plasma volume (adult): 3000 mL
role in the coagulation abnormalities after an HTR.
1650 mg Hb Several reports have suggested that red cell stroma
= 0.55 mg Hb/mL or 55 mg Hb/100 mL
3000 mL plasma could initiate DIC. Rabiner and Friedman107 showed
Barely visible hemolysis 15 mg Hb/100 mL that transfusion of autologous lysed dog RBCs led to
Plasma definitely pink 45 mg Hb/100 mL DIC. Birndorf and colleagues108 showed that transfu-
Plasma pink-red 60–100 mg Hb/100 mL sion of sonicated hemoglobin-free stroma into
From Masouredis SP, Chaplin H Jr: Transfusion management of autoimmune monkeys led to decreased factors I, II, V, and VIII and
hemolytic anemia. In Chaplin H Jr (ed): Immune Hemolytic Anemias. New platelets. Poskitt and coworkers109 suggested that the
York: Churchill Livingstone, 1985:177. ultra structure of red cell stroma resembles endotoxins
and that activation of the alternative complement
4 hours and increased progressively over 24 hours. pathway could occur when stroma are present in
These results suggested that antagonist production sufficient quantity.
might partly account for the variable pathophysio-
logic events seen in HTRs and AIHA.
Renal Failure Associated with HTRs
Disseminated Intravascular Coagulation Mollison and associates1 reported renal failure as
Associated with HTRs almost always being associated with complement acti-
vation during an HTR. Thus, it is usually found in
Disseminated intravascular coagulation (DIC), char- association with acute intravascular lysis, which is
acterized by thrombocytopenia, decreased factors I usually ABO mediated. Nevertheless, it has been
(fibrinogen), V, and VIII and the presence of fibrin described after DHTRs. Pineda and coworkers52
degradation products, sometimes occurs as the result found 17% percent of their patients with DHTRs to
of an HTR. Fortunately, it is rare, and it usually occurs have associated oliguria and 9% to have renal shut-
only after complement-mediated intravascular red down, but these patients all had serious underlying
cell destruction, which is usually associated with ABO disease, and in a later publication from the same insti-
incompatibility.1 Abnormal bleeding has been noted tution there were no cases of renal failure in 37 cases
many times after transfusion of ABO-incompatible of DHTRs.54 As with DIC, the exact relationship
blood.1 Indeed, blood bankers and anesthetists should between renal failure and HTR is unclear.
558 Immune Hemolytic Anemias

It is unclear from animal experiments whether VARIATION IN CLINICAL


hemoglobin or RBC stroma is involved in the renal SYMPTOMS ASSOCIATED
failure. It was first thought that hemoglobin is nephro- WITH ABO-INCOMPATIBLE HTRs
toxic, but that is no longer believed to be true, and
stroma itself does not seem harmful. Schmidt and
Holland110 transfused large quantities of compatible Although ABO incompatibility is by far the most
stroma without any observable reaction, but incom- common cause of fatal HTRs, and very small amounts
patible (e.g., ABO-incompatible) stroma led to severe of ABO incompatible blood can cause severe clinical
reactions (e.g., renal failure). symptoms, the range of symptoms seen among
It is now thought that the renal damage is an indi- individual patients is extraordinary. Considering
rect result of complement activation. The mechanisms how many patients still get transfused with ABO-
involved could be similar to those seen in renal incompatible blood, it is reassuring to note that only a
damage associated with immune complex disease, or small percentage of ABO-incompatible transfusions
those discussed previously for DIC. Anaphylotoxins are fatal.1 In one series of 40 patients receiving ABO-
could cause chemotaxis and subsequent enzyme incompatible blood, only four (10%) died.1 All four
release from leukocytes, thereby damaging renal patients had been transfused during or immediately
tissue, or there could be deposition of fibrin in the after surgery. Severe hypotension and DIC were the
renal microcirculation due to activation of the coagu- main clinical symptoms for all four patients.1 In two
lation cascade. older series describing 12 and 13 patients receiving
For renal failure after HTRs as for DIC, Capon and ABO-incompatible transfusions, none died or experi-
Goldfinger106 believe that cytokines might play an enced renal failure. Six patients received 50 mL of
important role. blood or less, but 11 of the 25 patients received 1 or
more full units of ABO-incompatible blood. Linden
and her colleagues113a reported on the outcome of 237
ABO-compatible transfusions in New York State; 47%
Treatment of HTRs of the patients had no obvious adverse effects. The
Usually, the only treatment necessary is to stop the five cases that follow show the wide range of clinical
causative transfusion, investigate its cause, and symptoms that can occur after ABO-incompatible
provide compatible blood for the next transfusion. If HTRs in individual patients.
the patient develops DIC or renal abnormalities,
urgent treatment becomes necessary. Shock and renal P A T I E N T 3 11 4 : A 21-year-old group O male re-
failure are the most common causes of death due to ceived 3 units of group A blood during surgery for spondy-
HTRs, and as hypotension is an important component lolisthesis. No unusual bleeding was noted; during surgery,
of both, this should be treated immediately by main- pulse, blood pressure, and urine output remained normal.
taining intravascular volume via the immediate infu- During the first 14 hours after surgery, his temperature rose
sion of colloids or crystalloids. Diuresis should be to 38.3°C but returned to normal the next day. No hemo-
induced as soon as possible to increase renal blood globin was visible in the plasma or urine. The DAT was
flow and to enable detection of renal shutdown. weakly positive. Before transfusion, the anti-A titer was
Mannitol (20 g as a 20% solution) was most commonly 128; it fell to 4 on the first day after surgery
used for this purpose for years, but more recently, and increased to 4096 at 10 days after surgery. Anti-A
furosemide (80–120 mg intravenously) has exceeded it hemolysins were not detected until day 10 (titer of 32).
in popularity. Urine output should be maintained for Survival of incompatible group A RBCs appeared to be
several hours at a minimum of 0.5 mL/kg/hour.1 good for 2 days after surgery, then started to disappear
Once oliguria is established, the patient should not be over the next 3 days; no A RBCs were detectable at 5
given more water than he or she can excrete (i.e., days. The patient did not become jaundiced, the urine was
500 mL/day). The patient’s renal function should be said to be of normal color; bilirubin, BUN, and creatine
monitored; if blood urea rises less than 20 mg/ remained normal from day 1 through day 91 after surgery.
dL/day, the patient usually responds well to conser-
vative treatment. Greater catabolic rates could suggest
that dialysis is necessary. Some workers have sug-
gested that dopamine is useful to combat shock and P A T I E N T 4 1 1 5 : A 70-year-old group B male
renal failure and that heparin and even exchange received 3 units of blood postoperatively; 1 unit was later
transfusion have been suggested as useful adjuncts to found to be group A. One hour after transfusion, the patient
the just-mentioned therapy.71,72,111-113 Treatment for had chills and fever as high as 40°C and experienced a
DIC has been well described in other publications and slight fall in blood pressure. The DAT was negative; no
will not be discussed here, other than to note that hemoglobin was obvious in the plasma. There was no
immediate rapid infusion of 50–100 mg of aqueous decline in hemoglobin, and bilirubin values were repeat-
heparin followed by the slow infusion of 250–350 mg edly normal during the week after the transfusion. The anti-
of heparin over the course of 24 hours has been A titer was 8 before transfusion and decreased to 2 after
recommended.111-113 the incompatible transfusion; on day 11, the titer was 128.
Hemolytic Transfusion Reactions 559

P A T I E N T S 5 A N D 6 11 6 : Patients 5 and 6 were Almost a quarter of the errors were a failure of blood
group B and group O, respectively; both received ABO- bank technology; for example, four cases (two imme-
incompatible blood. Patient 3, mistyped as group AB, diate HTRs and two DHTRs) were associated with
received 4 units of group AB during surgery and 8 units compatible crossmatches, and the remaining four
of AB+ and 18 units of group A blood during the next cases involved errors of interpretation or judgment.
week. Patient 4 received 2 units of group A during Honig and Bove120 found that 86.4% of the deaths due
surgery. Both patients had unexpected bleeding thought to to immediate (acute) HTRs were associated with ABO
be associated with DIC. The ABO incompatibility was not incompatibility; 9% were due to other antibodies (e.g.,
detected in Patients 3 and 4 until the seventh and fifth day, anti-c, -K, -M plus -P1, -S plus -c, or -P1); one death was
respectively, after surgery. Patient 3 died on day 28 after due to thermal hemolysis in a blood warmer, and in
surgery due to complications resulting from the HTR; one the cause was unclear. Two deaths were thought
Patient 4 died 7 days postoperatively of a myocardial to be due to DHTRs (one patient had anti-c and the
infarction, probably precipitated by the transfusion of other anti-c plus anti-E). Evaluation of the reactions
15 units of ABO-incompatible blood. due to non-ABO antibodies was difficult, as most of
the patients were moribund at the time of transfusion.
On analyzing similar records from the FDA,
P A T I E N T 7 117: A 92-year-old group A, Rh-negative Schmidt118 felt that many of the deaths reported to the
male with diverticulitis was mistyped as group AB because FDA as being due to HTRs were not proven as such,
of the use of a particular monoclonal anti-B. The hospital and in his analysis, he eliminated more than half of
did not detect anti-B in the patient’s serum. After a negative the cases and included those he felt were proven to be
antibody screen, blood was issued through an abbreviated directly attributable to transfusion. In Schmidt’s
cross-match (i.e., immediate-spin crossmatch). The patient analysis, 100% of the immediate reactions were due to
was given 3 units of group AB blood and 1 unit of group ABO antibodies.118 Of the 22 fatal immediate reactions
A blood, and no problems were reported. After the trans- due to ABO studied by Schmidt,118 17 (77%) were
fusion of a fourth unit of AB blood, the patient had a severe caused by the wrong patient being transfused, five
HTR, which resulted in renal failure and death (23%) by an error in test procedure labeling, and none
10 days later. After the transfusion reaction, the patient’s by errors in sample collection. Of 17 patients whose
pretransfusion RBCs were found to be group A with an deaths were due to misidentification, 12 deaths (71%)
acquired B antigen. The monoclonal anti-B used by the occurred in the operating room or intensive care unit,
hospital was formulated from the ES4 clone, which reacts none occurred in medical wards, and in five cases the
strongly with even weak acquired B antigens. A sample of location was not reported. Sixteen of the 22 patients
the patient’s serum taken before the transfusion reaction who died received 1 or fewer units of blood, six
was later found to contain a weak anti-B, detectable most received 2 units, and none received more than 2 units.
obviously by the antiglobulin test, which was not per- Nineteen of the 22 patients (82%) were group O and
formed at the crossmatch stage. The manufacturers of mon- received A or B blood.118
oclonal anti-B reagents prepared from the ES4 clone have In 1990, Sazama121 reviewed FDA data from 1976
since modified their reagents (i.e., lowered the pH) so that through 1985. During this period, 355 fatalities associ-
they now detect only the strongest examples of acquired B ated with transfusion were reported, 99 of which were
antigen (similar to other commercial anti-B). This case had excluded from further review because they were unre-
several unusual events coming together, but it serves to illus- lated to transfusion or were caused by transfusion-
trate that even a very weak ABO antibody (i.e., one not transmitted infection (e.g., hepatitis or acquired
detected by the immediate spin procedure) can cause a immune deficiency syndrome [AIDS]). Of the remain-
problem in a very sick patient. ing 256 reported deaths, 51% resulted from acute
hemolysis after the transfusion of ABO-incompatible
products. These deaths were due primarily to mana-
gerial, not clerical, errors. Other causes of death (in
order of frequency of report) included acute pul-
FATALITIES DUE TO HTRs monary injury (15%), bacterial contamination of
product (10%), delayed hemolysis (10%), damaged
Fortunately, fatalities due to HTRs are rare. product (3%), and graft-versus-host disease (0.4%). Of
Schmidt,118 Myhre,119 and Honig and Bove120 exam- the deaths due to ABO incompatibility, 85% involved
ined records of fatalities reported to the FDA for the transfusion of A, B, or AB RBCs to a group O recipient.
years 1976–1979 and found an incidence of about 1 Transfusion of group A RBCs to group O recipients
fatality per 500,000 transfusions. Honig and Bove120 accounted for 30% of all fatalities due to blood trans-
found that clerical errors were the major cause of the fusion. Nine deaths associated with non-ABO
fatal HTRs; they accounted for 89% of the errors, and immediate HTRs were attributed to anti-Jka, -Jka
in many cases, more than one person was involved. +anti-Jkb+Jk3, -Fya, or -E+K+P1. Of the 26 fatal
The single most common error was failure to ade- DHTRs, anti-c and/or anti-E were implicated in
quately identify the recipient prior to starting the 16 cases (62%). Antibodies in the Kidd system (anti-
transfusion; this accounted for 46% of the errors. Jka, -Jkb), Duffy system (anti-Fya, -Fyb, -Fy3), and Kell
560 Immune Hemolytic Anemias

system (anti-K, -Kpa) were associated with 10 (39%), (43% resulted solely from failure to identify the patient
8 (31%), and 7 (27%) of the DHTRs, respectively. As and/or unit prior to transfusion, and 11% resulted
16 of 26 patients (62%) had more than one antibody from phlebotomist error), while the blood bank was
specificity present in their sera, it was not clear responsible for 25% of errors and contributed, with
whether one or more of the specificities were the cause another hospital service, to 17% of the errors. Linden
of the fatal HTR. and coworkers123 considered that the risk of transfu-
Mummert and Tourault122 updated the FDA data sion of ABO-incompatible blood remains significant
gathered up to 1992. They concluded that nearly one and that additional precautions to minimize the likeli-
third of these fatalities could have been prevented by hood of such events should be considered.
adherence to proper procedure and reported that In 2000, Linden and Kaplan124 updated the New
transfusion of ABO-incompatible blood cells because York State findings through 1998. Erroneous adminis-
of error continues to be the primary cause of prevent- tration was observed for 1 of 19,000 RBC units admin-
able death. They identified failures in the following istered. Half of these events occurred outside of the
areas: blood bank (administration to the wrong recipient,
38%; phlebotomy errors, 13%). Isolated blood bank
1. Accurate identification of the patient errors, including testing of the wrong specimen, tran-
2. Recognition of the signs of a transfusion reaction and scription errors, and issuance of the wrong unit, were
appropriate action to discontinue the transfusion responsible for 29% of events. Many events (15%)
3. Verification that equipment in use was functioning involved multiple errors; the most common being
properly before and during use failure to detect at the bedside that an incorrect unit
4. Training of employees in adherence to standard had been issued.
operating procedures In 1996, the United Kingdom started a voluntary re-
porting system of major transfusion complications—
The investigators reported that failure to follow stan- the Serious Hazards of Transfusion (SHOT).125 For the
dard operating procedures was a significant problem, years 1996–1999, 54% of the reports (335 events) con-
and they advocated staff education, training, and mon- cerned the incorrect blood (97 of 335 events involved
itoring for adherence on an ongoing basis. They also ABO incompatibility) being given to the wrong
reported that failure to identify a reaction in progress patient. There were a total of 28 deaths due to trans-
contributed to many of the fatalities. They found that fusion and nine deaths suspected as being associated
most of the errors occurred outside of the blood bank with the transfusion. Four deaths were due (and two
and were largely in violation of existing operating pro- suspected of being due) to the incorrect components
tocols. They advocated facility comprehensive quality being transfused.125 Ibojie and Urbaniak126 performed
assurance programs to identify inappropriate proce- a retrospective study of transfusion errors in a large
dures in the facility and to reinforce the purpose and Scottish teaching hospital. Seventy-five percent of the
intent of required procedures. They also stressed the errors detected were classified as near misses. The
importance of the design of systems to prevent and number of patients transfused with the wrong blood
detect errors on an ongoing basis, and the importance was one in 27,007 units supplied. About 50% of the
of procedures for equipment validation. mistransfusion errors occurred at the patient’s bed-
side. The number of near misses was one in 9002. The
number of serious errors identified was one per
6752 units issued, or one per 2153 compatibility tests.
REPORTING OF ERRORS Fortunately, no fatal HTRs occurred. Callum and
AND NEAR MISSES colleagues127 performed a prospective study of
near misses using a no-fault medical-event report-
In New York State, significant incidents involving the ing system for transfusion medicine (MERS-TM).
collection, processing, or transfusion of blood must be Events and near-miss events (total 819) were recorded
reported. Linden and colleagues123 reviewed incident for a period of 19 months (median number, 51 per
reports received over a 22-month period involving month). No serious adverse patient outcome
transfusion of blood to other than the intended recipi- occurred, despite these events, with the transfusion of
ent, or release of blood of an incorrect group. Among 17,465 units of RBCs. Sixty-one events (7.4%) were
1,784,600 transfusions of red cell components, there potentially life threatening or could have led to
were 92 cases of erroneous transfusion (1 in 19,000) permanent injury (severity level 1). Of most concern
that met study criteria. There were 54 ABO-incompati- were three samples collected from the wrong patient,
ble transfusions (1 in 33,000); three of these (1 in 13 mislabeled samples, and 22 requests for blood
600,000) were fatal. Correction for under-reporting of for the wrong patient. Near-miss events were five
ABO-compatible errors resulted in an estimate of times more frequent than actual transfusion errors,
one per 12,000 as the true risk of transfusion error. and 68% of errors were detected before blood was
National application of New York State data results in issued. Sixty-one percent of events originated from
an estimate of 800–900 projected red cell-associated patient areas, 35% from the blood bank, and 4%
errors in the United States annually. The majority of from the blood supplier or other hospitals. Repeat
reported errors occurred outside of the blood bank collection was required for one in every 94 samples,
Hemolytic Transfusion Reactions 561

and one in 346 requests for blood components b. They compared the information on the patient’s
was incorrect. Education of nurses and alterations ID band with the blood component paperwork.
to blood bank forms were not by themselves effec- c. They compared the information on the blood
tive in reducing severe errors. An artifactual 50% component labels with the blood component
reduction in the number of errors reported was paperwork.
noted during a 6-month period when two chief 3. No deviations from policy were noted for handling
members of the event-reporting team were on tempo- of blood components outside of the blood bank,
rary leave. charting of required information, taking of vital
signs, use of intravenous solutions, duration of
Detection, Analysis, Frequency, transfusion, or follow-up of possible transfusion
and Prevention of Errors reactions.

Taswell and coworkers128 reviewed transfusion medi- An initial small audit revealed that variance from the
cine errors occurring at the Mayo Clinic from 1982 foregoing procedures was observed in three (50%) of
through 1992. They defined an error as any deviation six transfusions. The audit data initially demonstrated
from the standard operating procedure. Twenty- the following:
four standard operating procedures were monitored
for errors that related to donor processing, testing of 1. A systematic failure of transfusionists (mainly
donor blood, patient testing, and transfusion. The nurses, but some physicians and medical students)
estimate of the overall error rate and 95% confidence to identify patients properly before transfusion
interval fluctuated between 20 and 30 per 104 pro- 2. Sporadic variances in other transfusionists’ prac-
cedures. The transcription error rate declined from tices, such as the taking of vital signs and chart
21 to six per 104 procedures as a result of changes documentation
to systems using computer-generated labels and bar
codes. Several factors were found to cause variances from
Several investigators have audited errors occurring proper blood administration practices, including:
from when a unit of blood is first tested in the trans-
fusion service until it is transfused to the patient.129-131 1. Insufficient knowledge due to a deficiency in
This approach differs from the reports in the previous orientation or training in the procedure
section in that these are errors observed by an auditor 2. Behavioral or performance deficiency due to the
watching the process, rather than reported errors. It is lack of acceptance of the procedure, indifference to
likely that the audits are nearer to the truth than the the procedure, or carelessness
reported errors (e.g., FDA data). 3. System deficits
Most disturbing data has come from a Belgian
study. Baele and associates129 audited 3485 units of Even though all nurses are supposed to be orientated
blood transfused to 808 surgical patients. After the and trained in proper blood administration proce-
units left the blood bank, 165 errors were noted. dures, the audit process demonstrated knowledge
Fifteen of these errors were defined as major. Seven of deficits. Many nurses failed to perform the pretrans-
2772 units (0.25%, or one in 400) were transfused to fusion clerical cross-check properly because they felt
the wrong patient. Luckily, in six instances they pro- that they knew the patient so well that the clerical
voked no obvious symptoms; in one patient, a few mL checking was unnecessary. These nurses did not
of group B blood given to a group O patient caused realize that a blood bank error could result in issuance
chills and the transfusion was stopped. It is of concern of the wrong blood for their patient and that, without
to note that the episode was noted in the patient’s the clerical cross-check being done properly, their
notes, the unit was discarded, and the reaction was patient (whom they “knew so well”) might get blood
not reported to the blood bank. that was actually intended for someone else. As
Shulman and colleagues130,131 used a multidiscipli- transfusionists began to realize that one reason for
nary team to address this problem. The team con- doing the clerical cross-check was to catch a blood
sisted of representatives from the transfusion service, bank error, the hospital blood administration proce-
the administration, nursing, medical information dures won better acceptance. Furthermore, as each
services, and quality assessment coordinators from variance from proper blood administration practice
various clinical departments. Acceptable compliance was addressed, improvement occurred, and compli-
with institutional blood administration policies ance with pretransfusion clerical cross-checking
existed when the following were documented: approached 100%. In audits 126 to 175 (50 consecutive
transfusions), there was 100% compliance with blood
1. The intended recipient wore an ID band. administration policies. Other researchers have
2. Two licensed individuals did each of the following reported similar findings.126,132
while at the patient’s bedside: Shulman and Kent133 studied the incidence of unit
a. They compared the information on the patient’s placement errors, that is, the placing of RBC units in
ID band with the information on the blood com- the wrong section of the refrigerator. In a study of
ponent labels. 96,581 units at a large institution, they found an error
562 Immune Hemolytic Anemias

rate of 0.12% (112 units misplaced, one in 862), with patient (e.g., chart, beds, wall) and the practice of
about one third of these potentially leading to ABO- having nurses be responsible for initial wristband
incompatible transfusions if released (an ABO mis- placement.135
match error rate of 0.04% or 1 in 2610). They noted Lau and colleagues136 designed an improved wrist-
that placement errors are of concern because a band system. The wristband had the following special
crossmatch cannot always be counted on to detect features:
ABO incompatibility and usually will not detect Rh
incompatibility because most Rh-negative patients • Once attached, it cannot be removed except by
do not have anti-D. The investigators further noted cutting.
that placement errors are of great concern when • It has a pocket containing a transfusion label.
uncrossmatched blood is transfused in emergency sit- • A unique transfusion barcode is printed simultane-
uations. Their study showed the importance of fre- ously on each transfusion label and the correspon-
quent verification of unit placement to reduce the ding wristband by computer technology.
chance of an incompatible unit being released by • A transfusion label removed from the wristband
mistake.133 after attachment to the patient has a characteristic
tear-mark distinguishing it from one removed prior
Systems for Reducing Incidence of to attachment.
Transfusion of Blood to Wrong Patient
The blood bank accepted only those specimens
The most commonly used system involves iden- bearing the tear-marked transfusion labels. All blood
tification of the patient using a wristband. In a College units for this patient were labeled with this unique
of American Pathologists (CAP) Q-Probe study, transfusion code together with the patient’s details.
Renner and coworkers134 compared wristband iden- The nurses counter-checked the transfusion code on
tification errors for 712 hospitals. Phlebotomists the blood units against the transfusion code on the
checked patient wristbands on 2,463,727 occasions, patient’s transfusion wristband prior to transfusion.
finding 67,289 errors; in 33,308 instances, patient If the blood sample for compatibility testing was
wristbands were missing entirely. The median total drawn from the “wrong” patient, the intended
error rate was 2.2%; 10% of participants had error patient either did not carry a wristband or the trans-
rates of 10.9% or greater. Absent wristbands repre- fusion codes did not match at all. Pretransfusion com-
sented 49.5% of all errors; multiple wristbands patibility tests were performed on 2189 patient
with different information, 8.3%; wristbands with samples using this procedure, which was well
incomplete data, 7.5%; wristbands with erroneous accepted by both ward and blood bank staff. Two
data, 8.6%; wristbands with illegible data, 5.7%; and potential mismatched transfusions were avoided.
patients wearing wristbands with another patient’s These two clerical errors would not have been
identifying information, 0.5%. The monitoring for detected because neither patient had previous ABO
errors by phlebotomy staff was the most important grouping results.
policy associated with lower error rates. Initial place- Other approaches have been suggested. One sys-
ment of wristbands by nursing staff was the only tem consists of a coded locking system so that a blood
policy associated with increased error rates. They con- unit cannot be accessed without matching a three-
cluded that wristband identification error rates letter code that can be found only on the patient’s
depend on differences in hospital policy and proce- wristband. Several authors have reported on the
dure and should be responsive to quality improve- efficiency of this system.137-139 Jensen and Crosson140
ment efforts.134 described a system composed of the following
A follow-up study of 204 smaller hospitals com- components:
pared wristband errors.135 Phlebotomists examined
wristbands on 451,436 occasions and identified 25,800 1. A portable bedside scanner that reads barcoded
errors (total error rate, 5.7%). The absence of a wrist- patient and blood unit identification
band accounted for 64.6% of all errors reported; wrist- 2. A host computer system that accepts data from the
bands with missing information, 12.4%; multiple scanner
wristbands with different information, 12.1%; wrist- 3. Printed documentation of the transfusion
bands with erroneous information, 6.7%; illegible 4. Audit trail monitoring
wristbands, 3.5%; and patients wearing another
patient’s wristband, 0.7%. Factors found to correlate A commercial variation of this system, which is inte-
with lower error rates were the practice of sending grated with an automated blood typing system, has
written correspondence to the nursing service subsequently become available.141
involved for each error detected, the practice of Lumadue and colleagues142 felt that adherence to a
having nursing staff monitor wristbands on patient strict specimen-labeling policy would be an efficient
transfer, and laboratory accreditation from CAP. way to decrease errors leading to HTRs. Incorrectly
Factors found to correlate with higher error rates labeled specimens (rejected samples) were tested for
were the practice of allowing wristbands to be placed ABO and Rh type, and routine antibody screens were
on objects that could become separated from the performed. Test results were compared with historic
Hemolytic Transfusion Reactions 563

patient data or patient data obtained from subse- Patients Whose Sera Contain Antibodies
quently submitted (correctly) labeled specimens. For That Are Not Detected by Routinely
comparison, all discrepant serologic results from
appropriately labeled samples were also recorded.
Used Tests But Are Detectable by
Specimens that failed to meet the criteria for specimen Special Procedures
acceptance were 40 times more likely to have a blood There have been many reports in the literature of
grouping discrepancy. patients who had HTRs following transfusion of
blood that was compatible by saline, albumin, and
routine antiglobulin techniques, but alloantibody was
HEMOLYTIC TRANSFUSION demonstrable by techniques that are not used rou-
REACTIONS DUE TO ANTIBODIES tinely for cross-matching. In 1978, Snyder and
THAT ARE NOT DETECTABLE BY coworkers143 detected an anti-e that had caused an
ROUTINE PROCEDURES HTR but was detected only by an automated
Polybrene technique. Such antibodies rarely are
detectable by routine procedures. In 1982 and 1996,
Hemolytic transfusion reactions, both immediate and
Garratty and associates144,145 reported on more than
delayed, can occur without antibodies being detected
70 patients with HTRs but no detectable antibodies by
(at the time of the reaction) by routinely used tests.143-166
routine procedures. More than 70% presented with
These reactions appear to occur among three groups of
hemoglobinemia and hemoglobinuria. Sera were
patients:
tested using a manual Polybrene technique, polyeth-
ylene glycol (PEG), and ficin-treated RBCs (tube plus
1. Patients whose sera do not contain detectable anti-
capillary), and many were tested with increased
body at the time of the reaction but in which
amounts of patients’ sera, diluted sera, monocyte
detectable antibody appears later
monolayer assays, and enzyme-linked antiglobulin
2. Patients whose sera contain antibodies that are not
tests. Twelve of 70 antibodies (18%) (three anti-C,
detected by routinely used tests but are detectable
three anti-Jka, two anti-S, two anti-e, one anti-E, one
by special procedures
anti-Jkb), that were not detected by routine tests were
3. Patients whose sera appear to contain antibodies
detected by a Polybrene test; six of these 12 were
that are not detectable by routine procedures or
tested by PEG, and all reacted; three of the 12 reacted
multiple special procedures even after repeated
by a ficin-capillary technique. Three antibodies (one
transfusions and reactions
anti-e, one anti-C, one anti-Vel) became detectable by
routine tests after the reaction. Three e– and three C–
Patients Whose Sera Do Not Contain patients, with antibodies undetectable by any
Detectable Antibody at the Time of the methods, responded poorly to e+ and C+ RBCs but
Reaction But in Which Detectable responded well to e– and C– RBCs, respectively (sup-
ported by 51Cr RBC survival studies in three cases).
Antibody Appears Later Two other patients responded well to RBCs matched
Although the serology pattern for these patients is for several antigens (C, E, Jka, S) that the patients
similar to classic DHTR, transfused RBCs are lacked. The most common diagnoses of 67 patients
destroyed, sometimes rapidly, during the phase when were: leukemia (11 patients: six with chronic lympho-
antibody is undetectable. As early as 1957, Fudenberg cytic leukemia [CLL], five with other leukemias), mul-
and Allen58 described such patients. They performed tiple myeloma (six patients), lymphoma (five
51Cr RBC survival studies in three subjects who were patients), cancer (five patients), gastrointestinal bleed
known to have anti-s, -S, and -Jka, but at the time of the (five patients), and sickle cell disease (three patients).
study these were not detectable. The Cr-labeled s+, S+, One CLL patient who had repeated HTRs with no
and Jk(a+) RBCs had half-lives of 55 minutes, 9 hours, detectable antibodies was investigated for RBC
and 12 days, respectively. After the study, the anti-s destruction through an antibody-independent cellular
rose to a titer of 1, the anti-S rose to a titer of 8, and the mechanism.144 The patient’s lymphocytes were incu-
anti-Jka was still not detectable. The authors suggested bated in vitro with 51Cr-labeled autologous and
that there might be two main reasons for these findings. homologous RBCs in an NK cell cytotoxicity assay.
The first was that the antibody was present in the The results were noninformative.
serum at the time of the transfusion but the routine Maynard and associates146 also found Polybrene
techniques were not sensitive enough to detect it. The useful in detecting clinically significant antibodies not
second suggestion was that there was no antibody in detectable by routine procedures. A patient with
the serum but plenty in the spleen and/or other tissues. macroglobulinemia experienced chills, fever, hemo-
The pattern described by Fudenberg and Allen58 has globinemia, and hemoglobinuria after the transfusion
rarely been described in more recent literature, and it of one unit of RBCs shown to be compatible by a
could be that more modern serological procedures low-ionic-strength indirect antiglobulin (LIS-IAT)
detected such antibodies in the pretransfusion sera. The method. Serologic investigation was negative. Intra-
patterns described next seem to be the ones that are vascular hemolysis occurred with a second “compa-
observed in contemporary practice. tible” unit. Serologic tests were again negative by
564 Immune Hemolytic Anemias

LIS-IAT and ficin-AG methods but revealed anti-Jka experiment suggested that a factor (e.g., antibody) in
by the manual Polybrene technique. Both donors with Mrs. E’s serum was not responsible for the decreased
Jk(a+b–) and 51Cr studies of the second donor’s RBCs RBC survival observed in the original transfusion or
revealed a T1⁄ 2 of greater than 30 minutes with marked 51Cr studies.
intravascular hemolysis. RBCs from a C+, c+, Jk(a–) More recently, results of 51Cr RBC survival studies
donor that were compatible by LIS-IAT showed a 51Cr have suggested an antibody with defined blood
T1⁄ 2 of 100 minutes with slight intravascular lysis. Four group specificity as the cause of HTR. Davey and
transfusions of Jk(a–), C– blood were uneventful, but associates153 described a patient who had HTR with
5 days later the patient’s hemoglobin declined. The no detectable antibody. The patient had a severe
following day, anti-E was demonstrable exclusively DHTR (hemoglobinemia and hemoglobinuria) 10
by the Polybrene test. 51Cr-labeled Jk(a–), C–, E– RBCs days after transfusion of 4 units of blood. No anti-
had normal 24-hour survival. The patient’s hemoglo- bodies were detected by multiple serological proce-
bin rose to 11 g/dL after transfusions of Jk(a–), C–, dures, either routine or special. Following red cell
E– RBCs, and he was discharged. phenotyping, it was ascertained that the only antigen
Others have reported similar findings.147,148 that the patient lacked and the units possessed was c.
Although Polybrene appeared to be the most efficient 51Cr RBC survival studies showed that c-positive
test to detect these unusual antibodies, it might not be RBCs had 48% survival at 3 hours and less than 1%
the best for routine compatibility test procedures. survival at 24 hours, compared with 93% survival at
Lown and Willis149 tested random antibodies, which 3 hours and 80% at 24 hours for c-negative RBCs.
were detected by the Polybrene test but negative by Eight c-negative units were transfused with no prob-
IAT, by a monocyte monolayer assay and found that lems; no anti-c was detectable at the time of transfu-
many of these would be predicted to be clinically sion or at 2 months or 6 months after transfusion.
insignificant. Baldwin and colleagues154 described a similar case.
Serological studies using multiple techniques
demonstrated only an anti-Bga; these studies
Patients Whose Sera Appear to Contain included both standard procedures and more sensi-
Antibodies That Are Not Detectable by tive experimental techniques. A 51Cr survival study,
Routine Procedures or Multiple Special using RBCs from a random unit compatible in vitro
Procedures Even After Repeated with conventional techniques, showed 72% survival
at 1 hour and 7% survival at 24 hours. R2R2 (e-nega-
Transfusions and Reactions tive) RBCs in a second 51Cr survival study showed
As early as 1959, Stewart and Mollison150 and in 1961, 90% survival at 1 hour and 92% survival at 6 hours.
Kissmeyer-Nielsen and colleagues151 described The patient was transfused with R2R2 units, which
patients who had HTRs after serologically compatible were tolerated well and survived normally. Extensive
blood. 51Cr RBC survivals were grossly abnormal. serologic testing still demonstrated only an anti-Bga.
Chaplin and Cassell80 followed a patient with SCD A third 51Cr survival study, 10 months after the
who had HTRs and no demonstrable alloantibodies first study, with an R1R1 (e-positive) sample showed
for 2 years, and they performed 17 RBC survival 90% survival at 1 hour and 42% survival at 6 hours.
studies. Survival of transfused RBCs in this patient A fourth study, using a larger aliquot of R2R2
varied from a half-life of less than 1 day to a normal (e-negative) 51Cr-labeled RBCs, examined over the
survival of 30 days; the 30-day RBC survival was course of 2 weeks, showed a near-normal 21-day sur-
obtained on blood from the patient’s sister; therefore, vival of 50%. These 51Cr survival studies, along with
it did appear that the patient had an antibody with a normal survival of e-negative units, suggest that this
specificity that was not apparent from the RBC sur- patient destroyed e-positive RBCs despite negative
vival studies. van Loghem and coworkers152 also serologic testing.154
showed rapid destruction of 51Cr-labeled red cells in Garratty and colleagues described two further
five patients who had HTRs with no detectable anti- patients for whom blood was selected on the basis of
body (half-lives varied from 3 hours to 14 days). The the patients’ phenotypes. Anti-C and anti-e, respec-
authors felt that this phenomenon was not due to cir- tively, were suspected because of proven destruction
culating antibodies. This conclusion was based on the of RBCs having the appropriate antigen and survival
results of an interesting experiment. They incubated of RBCs lacking the antigen. These antibodies were
RBCs from a donor (Mrs. G) in the serum of a previ- undetectable by all tests in our lab and those of several
ous recipient (Mrs. E). Mrs. G’s RBCs had been shown specialist labs. RBC survival was studied in one case
previously to be destroyed rapidly in recipient Mrs. E. with 51Cr-labeled RBC and in the other case by differ-
Following incubation in vitro, the RBCs were labeled ential agglutination. One patient with CLL, who had
with 51Cr and reinjected back into Mrs. G; the RBCs repeated HTR with no detectable antibodies, was
survived normally. This experiment was performed investigated for RBC destruction through a nonanti-
twice: first with serum taken 6 months after the origi- body-dependent cellular mechanism. Patient’s lym-
nal transfusion of “incompatible” blood and the phocytes were incubated in vitro with 51Cr-labeled
second time with serum taken 3 weeks after the trans- autologous and homologous RBCs (NK cell cytotoxic-
fusion; RBC survival was normal both times. This ity assay). The results were noninformative.144,155
Hemolytic Transfusion Reactions 565

Harrison and coworkers156 described an HTR Mollison and coworkers161 criticized the inter-
caused by anti-C that was not detectable by any sero- pretation of the 51Cr survival curves used by Panzer
logical procedures, including enzymes, but which was and colleagues.160 The method used by the latter
defined by 51Cr studies; the HTR was associated with researchers was meant to be applied to the survival of
hemoglobinemia and hemoglobinuria. autologous RBCs, not to potentially incompatible
RBCs. When the RBC survival curve has more than
Can HLA Antibodies Cause HTRs? one component to the method, it is not applicable. On
reinterpreting the data, Mollison and coworkers161 felt
HLA antibodies are usually not considered important that the 51Cr survival curves published by Panzer and
with regard to RBC survival. Bg antibodies (anti-HLA- colleagues160 show that the T50Cr in their cases
B7, -17, -28) have been shown not to usually destroy 3 through 6 was approximately 24–30 days (i.e., prob-
reactive RBCs. The antibodies would not be expected to ably within normal limits). Cases 1 and 2 showed
cause many RBC problems because there are very few about 25% destruction of the remaining RBCs in the
HLA antigens on mature RBCs. On the other hand, IgG first 24 hours.
HLA antibodies are known to activate complement In 1993, Weitekamp and associates162 reported very
efficiently and perhaps, on occasion, they could cause convincing data that HLA might sometimes cause
shortened RBC survival. There are some reports in the HTRs. A woman with gastrointestinal bleeding (one
literature supporting this hypothesis.157-163 van der unit RBCs per day), received 13 uncomplicated RBC
Hart and associates157 performed 51Cr RBC survival transfusions in a 2-week period. With the 14th unit of
studies in a patient with lymphocytotoxic antibodies to RBCs, she developed shaking chills, nausea, vomiting,
HLA-B40, -B13, and -B7, with RBCs from a donor and red urine. LDH was 2024 IU/L and haptoglobin
with strong RBC HLA antigens (A2; B7, 40). About 60% less than 5 mg/dL, with a disproportionate decrease
of the RBCs were removed, with a T50Cr of about in hemoglobin. Similar reactions were seen with 3 of
100 minutes, and the rest with a T50Cr of 20 hours. the next 6 units (2 of which were saline washed).
Nordhagen and Aas158 studied a patient whose serum Postreaction blood samples were grossly hemolyzed
reacted with RBCs from HLA-B28–positive donors. A and, despite enhancement, they contained no RBC
small component (20%) of the incompatible RBCs had alloantibodies other than the previously recognized
a reduced survival (T50Cr of 1.5 days), but the main anti-D and -Fyb. The patient’s HLA type was A1,26;
component had normal survival. B44,70. Potent HLA antibodies specific for private A2
Panzer and colleagues159 showed that 51Cr-labeled and public A2-B17, A2-28, and A2-28-9 were demon-
RBCs that were incompatible with HLA antibodies strated by adsorption and elution lymphocytotoxic
present in the sera of six women had shortened sur- assays. The patient subsequently received 10 units
vival in every case, especially when anti-HLA-B7 was from HLA compatible donors (6 new) without
involved. In further studies, Panzer and colleagues160 difficulty. Antibodies reacting 1+ in IAT against HLA-
performed a prospective study to see whether HLA incompatible RBCs were detected 3.5 weeks after
sensitization is associated with increased RBC hemolysis began. These antibodies reacted with each
destruction after HLA-incompatible transfusion; of the four donors implicated in the transfusion reac-
51Cr-labeled RBC survival and site of sequestration tions and were negative with RBCs of nine donors (13
were monitored in nine patients in whom HLA anti- units) that were tolerated. HLA typing and AHG
bodies had developed after RBC transfusion. The white-cell cross-matching showed that all four donors
donors selected were compatible in ABO- and RBC- implicated in hemolysis were incompatible with the
specific antigen systems but were mismatched for the HLA antibody; whereas the nine donors of tolerated
HLA antigen in question. Hemolytic transfusion units were HLA compatible. The patient died 2 weeks
reactions occurred in all four patients who received after total gastrectomy, of surgical complications. This
HLA-B7–incompatible RBCs. A direct radioimmune appears to be first reported case of repeated, severe,
anti-IgG test became positive, 51Cr RBC survival was symptomatic hemolytic reactions after transfusion of
very short, and excess sequestration in the spleen was HLA-incompatible RBCs. The high titer (128) of the
measured. There was a rise in serum lactic dehydro- lymphocytotoxic antibody might explain these
genase and a fall in haptoglobin. HLA-B7 antibody unusual reactions. The authors suggested that HLA-
was detected in the eluate prepared from RBCs col- reactive alloantibodies should be investigated in
lected after transfusion. A similar reaction was found patients with unexplained hemolytic transfusion reac-
in only one further patient, caused by an HLA-A2 tions.162 A similar case was described by Benson and
incompatibility. No indications of immune-medicated associates.163
RBC sequestration were discernable after transfusion
of HLA-B7 compatible RBC in one of the patients who NONHUMORAL MECHANISMS
had shown a reaction with HLA-B7 incompatible
blood, nor in any of the other patients who received It is possible that the RBCs are being destroyed by an
HLA-B7–compatible RBCs. The hemolytic transfusion antibody-independent, cell-mediated mechanism. For
reactions could not be anticipated by conventional instance, NK cells are known to destroy other cells by
cross-match procedures, nor by the measurement of such a mechanism. Garratty164 suggested this as a
the 51Cr survival 1 hour after transfusion. cause of HTRs as early as 1981 but was unable to
566 Immune Hemolytic Anemias

prove it; an NK cell assay was performed using the and anti-A,B are almost always transferred. Two
donor RBCs and mononuclear cells from a patient major events prevent passively acquired ABO anti-
with chronic lymphocytic leukemia who had HTRs bodies from causing a clinical problem. First, the anti-
with no detectable antibodies.144,164 It is interesting to bodies are, of course, diluted in vivo. The average
note that Gilsanz and colleagues165 have shown in one anti-A titer is about 128; thus, if no other factors play
case that NK cells can cause “DAT-negative” AIHA. a role, a small adult—for instance, a 5-ft, 100-lb
Thus, the hypothesis is worth retaining. woman with a plasma volume of 1200 mL—would
need to receive only about 10 mL of plasma to theo-
retically have detectable antibody.167,168 In practice,
HTRs ASSOCIATED WITH PASSIVELY the antibody probably would not be detectable on the
TRANSFUSED ALLOANTIBODIES patient’s RBCs or in the serum because ABH antigens
are distributed widely in the recipient, and much of
The problem of passively acquired alloantibodies is an the passively transfused antibody is inhibited by ABH
old one. Because group O blood was first transfused blood group substances present in the recipient’s
to patients who were not group O, clinicians and plasma and tissues. Passively acquired ABO anti-
immunohematologists have had to deal with the pas- bodies are usually detected in recipients only
sive transfer of anti-A, anti-B, and anti-A,B to group when the donor antibody is of high titer, and
A, B, or AB recipients. As more plasma products (e.g., hemolytic transfusion reactions occur only when the
platelets, fresh-frozen plasma, coagulation factors) plasma contains antibody of exceptionally high titer,
were used for recipients who were sometimes not when large volumes of plasma are transfused, or
ABO identical, the problems grew. With the advent of when young children or infants receive transfu-
bone marrow transplantation (BMT), the increasing sions.1,169-171 Mollison and coworkers1 reviewed early
use of intravenous immune gamma globulin (IVIgG), work in which 250 mL of group O plasma was delib-
and more recently, the use of intravenous (IV) anti-D erately transfused to group A volunteers. In two
(an Rh[D] immune globulin) in Rh-positive recipients, studies, the lowest titer of anti-A agglutinins associ-
new problems have arisen.166 ated with in vivo hemolysis were 512 and 640; 40%
The amount of plasma contained in a unit of RBCs and 23%, respectively, of group O donors were found
will be influenced by the hematocrit of the donor and to have anti-A in such titers. Thousands of units of
how that unit was prepared. (The lowest acceptable whole blood from “safe” (ABO-antibody titers <200)
hematocrit is 38%; there is no mandated upper value, group O donors were transfused to group A and B
but the highest normal male hematocrit is around 54%, recipients in the Korean and Vietnam wars with very
the hematocrit must be adjusted to take into account few ill effects.172,173 Schwab and associates174 per-
the amount of anticoagulant present—63 mL for a formed a 2-year prospective study using group O
450 mL unit and 70 mL for a 500 mL unit.). Blood cen- uncross-matched “packed” RBCs for trauma cases
ters use a “soft spin” (e.g., 2200 rpm for 7 minutes in a and found no hemolytic transfusion reactions after
Sorvall RC3 centrifuge) when preparing platelet prod- 880 transfusions.
ucts from the unit, and a “hard spin” (e.g., 4000 rpm From a practical point of view, it is usually advised
for 6 minutes in a Sorvall RC3 centrifuge) when only to continue transfusing group O (rather than the
plasma is taken off. The resulting hematocrits of the patient’s own type-specific blood) to a group A or B
“packed” RBCs before adenine-saline solution (e.g., patient who has had to receive group O blood, until
Adsol, Fenwal Laboratories, Roundlake, IL) is added, anti-A or -B is no longer detectable (at 37ºC or by
are, on average, 81% and 93%, respectively, at the antiglobulin test) in the patient’s serum.166
American Red Cross in Los Angeles. Recently, 500 (±
50 mL) units are being collected, in addition to 450 (± PASSIVE TRANSFER OF RBC ALLOANTIBODIES
45 mL) units; this, too, will influence the amount of OTHER THAN ABO
residual plasma. Taking all those factors into account,
the amount of plasma remaining in “packed” RBCs Standard 5.8.3.1 of the 22nd edition of the American
can be calculated to be 12–40 mL.166 As the increasing Association of Blood Banks (AABB)175 states that only
trend is for blood centers to remove as much plasma as serum or plasma from donors with a history of transfu-
possible from RBC products (e.g., in the Los Angeles sion or pregnancy should be tested for unexpected RBC
Blood Center, two thirds of the “packed” RBCs, before antibodies, but most blood centers find it more conven-
addition of adenine-saline solution, have a hematocrit ient to screen all donors. Thus, it is rare for non-ABO
of about 90%; after adding adenine-saline solution, the alloantibodies to be transferred passively from donor to
hematocrit is 55–65%), the amount of residual plasma recipient. As DATs are not performed routinely, RBC-
is often at the lower end of the range.166 bound autoantibodies can be transferred to the recipi-
ent. Such autoantibodies probably have little clinical
TRANSFUSION OF GROUP O BLOOD significance to the recipient if they are causing no prob-
TO A, B, OR AB RECIPIENTS lems (i.e., hemolytic anemia) in the donor. Only about
0.2% of blood donors have 37ºC-reactive allo-
When group O blood (even “packed” RBCs) is trans- antibodies176; this frequency is obviously dependent on
fused to group A, B, or AB individuals, anti-A, anti-B, the techniques used. Even if alloantibodies were
Hemolytic Transfusion Reactions 567

present in a unit of blood, they would have to be excep- donors (who are not group AB) who do not have an
tional antibodies (i.e., of a high enough titer to with- identical ABO type could have passively acquired
stand dilution by recipient plasma) to cause any ill anti-A,B and/or anti-A, and/or anti-B. As discussed
effects in the recipient. High-titer donor alloantibodies previously for transfusion of group O blood,
can cause positive DATs and perhaps even be hemolytic anemia rarely ensues, but as larger volumes
detectable in the recipient’s plasma, but it is still of plasma are often involved (i.e., apheresis products),
unlikely that they would cause a significant reaction in there is an increased risk of problems. These problems
the recipient. This statement is supported by the might involve only a positive DAT due to sensitiza-
paucity of reports of any such reactions in the literature tion of the recipient’s RBCs with anti-A or -B. This can
and is emphasized by the minimal clinical ill effects confuse the investigator, as eluates are routinely
encountered when D-positive volunteers are transfused tested against only group O RBCs. Before performing
(sometimes deliberately) with plasma containing anti- extensive workups (e.g., evaluating drug-dependent
D, anti-C, or anti-K,177-179 and by the current practice of antibodies as a cause of a positive DAT), it is always
injecting powerful anti-D into D-positive patients with wise to ask whether the patient has received platelets
autoimmune thrombocytopenic purpura.180 from a donor of different ABO type. On occasions, the
The literature includes several reports of donor passively transfused ABO antibodies cause hemolytic
antibodies reacting with another transfused unit anemia, and on rare occasions this has been fatal.190-204
(“interdonor incompatibility”)181-187; most of these Shanwell and colleagues201 reported that nine of 11
were associated with anti-K. The majority of these group A recipients developed a positive DAT after
reactions were mild (e.g., fever, chills, increased receiving platelet concentrates (five had IgG + C3d,
bilirubin), but three were quite severe.182,183,186 Some and four had only C3d on their RBCs); none of the
of the reactions occurred before donor screening was nine patients showed signs of hemolysis.
common practice, and two were associated with anti- Coagulation Factors. Cryoprecipitate, factor VIII,
K that were not detected by an automated enzyme and factor IX products have all caused immune
technique.181,186 It is unknown whether these antibod- hemolysis in patients.205-212 The hemolysis was due in
ies would have caused similar reactions if transfused all cases to passively transferred ABO antibodies.
to a K-positive recipient. The association of such reac- Mild anemia is common among hemophiliacs, and it
tions with interdonor incompatibility is probably has been suggested that immune hemolysis caused by
because there would be much more antibody on each passively acquired antibodies might be a cause of the
K-positive RBC when only 1 unit of RBCs (i.e., the K- anemia; Buchanan and associates211 presented data
positive donor unit) is K positive. that did not support this hypothesis. With the advent
Naczek and colleagues188 found only minimal sero- of more highly purified and recombinant therapeutic
logic and no clinical problems when patients were coagulation factors, problems with passively trans-
transfused with RBCs from units containing alloanti- ferred ABO antibodies should become even rarer.
bodies. They evaluated 42 patients transfused with
RBCs from units containing anti-K, anti-D, anti-E, anti-
C, anti-C, anti-Fya, anti-Jka, and anti-A1. Antibodies LABORATORY INVESTIGATION OF HTRs
were detectable in a serum sample from only one recip-
ient; this patient had received 3 units containing anti-E The Standards of the AABB175 suggest that only three
and/or anti-Fya. One patient had a positive DAT with tests are mandated to exclude an immune etiology for
anti-D in an eluate from the RBCs. No clinical evidence an HTR. These simple tests are a repeat ABO group, a
of hemolysis was observed in any patient. DAT and visual examination of the recipient’s plasma
Combs and coworkers189 (from Duke University) for hemoglobin. The rationale for this is the finding,
transfused 253 units of RBCs, from donors who had by the Mayo Clinic, that approximately 90% of recip-
alloantibodies, to 187 random patients. Only 10% of ients have a positive DAT after an HTR.55,56 The
these recipients had antibody detectable in their patients who do not have a positive DAT and do have
serum after transfusion (all detectable antibodies were definite signs of a HTR are usually those associated
anti-D or C + D). No HTRs were reported. The anti- with ABO incompatibility. In ABO incompatibility, all
body-containing units were obtained at a discounted the sensitized RBCs might have been removed from
price. The authors concluded that large-scale use of the circulation. In such a case, hemoglobinemia,
RBC units from donors with alloantibodies is safe and obvious by visual inspection of the plasma, will
likely to have minimal impact on a busy transfusion always be present. Table 14-9 shows that destruction
service. of 5 mL of RBCs will lead to an obviously pink
plasma (especially if compared with pretransfusion
plasma), and even destruction of as little as 2 mL of
PASSIVE TRANSFER OF RBC ALLOANTIBODIES RBCs might be detectable visually.
IN PLATELET, GRANULOCYTE, PLASMA, Many investigators perform more than these three
AND COAGULATION PRODUCTS tests. Others might include repeat ABO and Rh typing,
antibody screening, and cross-matching. As mentioned
Platelets and Granulocytes. Any group A, B, or AB earlier, sometimes all the routine serological tests are
patient receiving platelets or granulocytes from nonproductive, and if the evidence for an HTR is good,
568 Immune Hemolytic Anemias

special tests may be performed. These might include 23. Singer ST, Wu V, Mignacca R, et al: Alloimmunization and
different antibody potentiators (e.g., enzymes, PEG, erythrocyte autoimmunzation in transfusion-dependent tha-
lassemia patients of predominantly Asian descent. Blood
Polybrene), different techniques (gel, solid phase), dif- 2000;96:3369–3373.
ferent antiglobulin sera (anti-C3, -IgA, -IgM), different 24. Ho H-K, Ha S-Y, Lam C-K, et al: Alloimmunization in Hong
antibody-antigen ratios (increased volumes of serum), Kong southern Chinese transfusion-dependent thalassemia
and most important, use of another laboratory (prefer- patients. Blood 2001;97:3999–4000.
25. Orlina AR, Unger PJ, Koshy M: Post-transfusion alloimmu-
ably a Reference Laboratory). nization in patients with sickle cell disease. Am J Hematol
1978;5:101–106.
26. Davies SC, McWilliam AC, Hewitt PE, et al: Red cell alloim-
R E F E R E N C E S munization in sickle cell disease. Br J Haematol 1986;
63:241–245.
1. Mollison PL, Engelfriet CP, Contreras M: Blood Transfusion in 27. Sarnaik S, Schornack J, Lusher JM: The incidence of develop-
Clinical Medicine, 10th ed. Oxford: Blackwell Scientific, 1997. ment of irregular red cell antibodies in patients with sickle cell
2. Grobbelaar BG, Smart E: The incidence of isosensitization anemia. Transfusion 1986;26:249–252.
following blood transfusion. Transfusion 1967;7:152. 28. Ambruso DR, Githens JH, Alcorn R, et al: Experience with
3. Grove-Rasmussen H: Routine compatibility testing. Trans- donors matched for minor blood group antigens in patients
fusion 1964;4:200–205. with sickle cell anemia who are receiving chronic transfusion
4. Tovey GH: Preventing the incompatible blood transfusion. therapy. Transfusion 1987;27:94–98.
Haematologia 1974;8:171–176. 29. Reisner EG, Kostyu DD, Phillips G, et al: Alloantibody
5. Spielmann W, Seidl S: Prevalence of irregular red cell anti- responses in multiply transfused sickle cell patients. Tissue
bodies and their significance in blood transfusion and antena- Antigens 1987;30:161–166.
tal care. Vox Sang 1974;26:551–559. 30. Koshy M, Burd L, Wallace D, et al: Prophylactic red-cell trans-
6. Walker RH, Lin D-T, Hartrick MB: Alloimmunization follow- fusions in pregnant patients with sickle cell disease: A ran-
ing blood transfusion. Arch Pathol Lab Med 1989;113:254–260. domized cooperative study. N Eng J Med 1988;319:1447–1452.
7. Hewitt PE, Macintyre EA, Devenish A, et al: A prospective 31. Cox JV, Steane E, Cunningham G, et al: Risk of alloimmuniza-
study of the incidence of delayed haemolytic transfusion tion and delayed hemolytic transfusion reactions in patients
reactions following peri-operative blood transfusion. Br J with sickle cell disease. Arch Intern Med 1988;48:2485–2489.
Haematol 1988;69:541–544. 32. Luban NL: Variability in rates of alloimmunization in differ-
8. Hoeltge GA, Domen RE, Rybicki LA, Schaffer PA: Multiple ent groups of children with sickle cell disease: Effect of ethnic
red cell transfusions and alloimmunization. Arch Pathol Lab background. Am J Pediatr Hematol Oncol 1989;11:314–319.
Med 1995;119:42–45. 33. Vichinsky EP, Earles A, Johnson RA, et al: Alloimmunization
9. Heddle NM, Soutar RL, O’Hoski PL, et al: A prospective in sickle cell anemia and transfusion of racially unmatched
study to determine the frequency and clinical significance of blood. N Eng J Med 1990;322:1617–1621.
alloimmunization post-transfusion. Br J Haematol 1995;91: 34. Rosse WF, Gallagher D, Kinney TR, et al: Transfusion and
1000–1005. alloimmunization in sickle cell disease. Blood 1990;76:
10. Lostumbo MM, Holland PV, Schmidt PJ: Isoimmunization 1431–1437.
after multiple transfusions. N Eng J Med 1966;275:141. 35. Wayne AS, Kevy SV, Nathan DG: Transfusion management of
11. Perkins HA: Isoantibodies following open heart surgery. sickle cell disease. Blood 1993;81:1109–1123.
In Hollander LP (ed): Proceedings of the 11th Congress 36. Tahhan HR, Holbrook CT, Braddy LR, et al: Antigen-matched
International Society for Blood Transfusion, Sydney 1966; donor blood in the transfusion management of patients with
Bibl. haemat. No. 29, Part 3. Basel: Karger, 1968:831. sickle cell disease. Transfusion 1994;34:562–569.
12. Blumberg N, Peck K, Ross K, Avila E: Immune response to 37. Garratty G: Severe reactions associated with transfusion of
chronic red blood cell transfusion. Vox Sang 1983;44:212–217. patients with sickle cell disease. Transfusion 1997;37:357–361.
13. Ting A, Pun A, Dodds AJ, Atkinson K, Biggs JC: Red cell 38. Wallhermfechtel MA, Pohl BA, Chaplin H: Alloimmunization
alloantibodies produced after bone marrow transplantation. in patients with warm autoantibodies: A retrospective study
Transfusion 1987;17:145–147. employing three donor alloabsorptions to aid in antibody
14. Brantley SG, Ramsey G: Red cell alloimmunization in multi- detection. Transfusion 1984;24:482–485.
transfused HLA-typed patients. Transfusion 1988;28:463–466. 39. Issitt PD, Combs MR, Bumgarner DJ, et al: Studies of antibod-
15. Ramsey G, Cornell FW, Hahn LF, et al: Red cell antibody prob- ies in the sera of patients who have made red cell autoanti-
lems in 1000 liver transplants. Transfusion 1989;29:396–400. bodies. Transfusion 1996;36:481–486.
16. Fluit CRMG, Kunst VAJM, Drenthe-Schonk AM: Incidence of 40. Leger RM, Garratty G: Evaluation of methods for detecting
red cell antibodies after multiple blood transfusions. alloantibodies underlying warm autoantibodies. Transfusion
Transfusion 1990;30:532–535. 1999;39:11–16.
17. Economidou J, Constantoulakis M, Augoustaki O, Adinolfi M: 41. James P, Rowe GP, Tozzo GG: Elucidation of alloantibodies in
Frequency of antibodies to various antigenic determinants in autoimmune haemolytic anaemia. Vox Sang 1988;54:167–171.
polytransfused patients with homozygous thalassaemia in 42. Laine ML, Beattie KM: Frequency of alloantibodies accompa-
Greece. Vox Sang 1971;20:252–258. nying autoantibodies. Transfusion 1985;25:545–546.
18. Coles SM, Klein HG, Holland PV: Alloimmunization in two 43. Morel PA, Bergren MO, Frank BA: A simple method for the
multitransfused patient populations. Transfusion 1981;21:462. detection of alloantibody in the presence of warm autoanti-
19. Sirchia G, Zanella A, Parravicini A, et al: Red cell alloantibod- body [abstract]. Transfusion 1978;18:388.
ies in thalassemia major. Transfusion 1985;25:110–112. 44. Sokol RJ, Hewitt S, Booker DJ, et al: Patients with red cell
20. Michail-Merianou V, Pamphili-Panousopoulou L, Piperi- autoantibodies: Selection of blood for transfusion. Clin Lab
Lowes L, Pelegrinis E, Karaklis A: Alloimmunization to red Haematol 1988;10:257–264.
cell antigens in thalassemia: Comparative study of usual 45. Issitt PD, Anstee DJ: Applied Blood Group Serology, 4th ed.
versus better-match transfusion programmes. Vox Sang Durham, NC: Montgomery Scientific, 1998:974.
1987;52:95–98. 46. Garratty G: Clinical significance of antibodies reacting opti-
21. Spanos T, Karageorga M, Ladis V, et al: Red cell alloantibodies mally at 37ºC. In Clinically significant and insignificant anti-
in patients with thalassemia. Vox Sang 1990;58:50–55. bodies. Washington, DC: American Association of Blood
22. Prati D: Benefits and complications of regular blood trans- Banks, 1979:29–49.
fusion in patients with beta-thalassaemia major. Vox Sang 47. Mollison PL: Blood-group antibodies and red-cell destruction.
2000;79:129–137. Br Med J 1959;2:1035–1130.
Hemolytic Transfusion Reactions 569

48. Giblett ER: Blood group alloantibodies: An assessment of (SCD): Evidence for autologous RBC destruction [abstract].
some laboratory practices. Transfusion 1977;17:299–307. Transfusion 1993;33(Suppl.):22S.
49. Issitt PD: Antibodies reactive at 30 centigrade, room tempera- 72. Friedman DR, Kim HC, Manno CS: Hyperhemolysis associ-
ture, and below. In Clinically significant and insignificant anti- ated with red cell transfusion in sickle cell disease [abstract].
bodies. Washington, DC: American Association of Blood Transfusion 1993;33(Suppl.):14S.
Banks, 1979:13. 73. Cullis JO, Win N, Dudley M, Kaye T: Post-transfusion hyper-
50. Mollison PL: The clinical significance of red cell alloantibodies haemolysis in a patient with sickle cell disease: Use of steroids
(and autoantibodies) in blood transfusion. In Polesky HF, and intravenous immunoglobulin to prevent further red cell
Walker RH (eds): Safety in Transfusion Practices. Skokie, IL: destruction. Vox Sang 1995;69:355–357.
College of American Pathologists, 1982:131. 74. Syed SJ, Sears DA, Werch JB, et al: Case reports: Delayed
51. Garratty G: Predicting the clinical significance of alloantibodies hemolytic transfusion reactions in sickle cell disease. Am J
and determining the in vivo survival of transfused red cells. In Med Sci 1996;312:175–181.
Clinical and serological aspects of transfusion reactions. 75. King KE, Shirey RS, Lankiewicz MW, et al: Delayed hemolytic
Washington, DC: American Association of Blood Banks, 1982:91. transfusion reactions in sickle cell disease: Simultaneous
52. Pineda AA, Brzica SM, Taswell HF: Hemolytic transfusion destruction of recipients’ red cells. Transfusion 1997;37:376–381.
reaction: Recent experience in a large blood bank. Mayo Clin 76. Petz LD, Calhoun L, Shulman IA, et al: The sickle cell hemo-
Proc 1978;53:378–390. lytic transfusion reaction syndrome. Transfusion 1997;37:
53. Pineda AA, Taswell HF, Brzica SM: Transfusion reaction: An 382–392.
immunologic hazard of blood transfusion. Transfusion 77. Aygun B, Padmanabhan S, Paley C, et al: Clinical significance
1978;18:1–7. of RBC alloantibodies and autoantibodies in sickle cell patients
54. Moore SB, Taswell HF, Pineda AA, et al: Delayed hemolytic who received transfusions. Transfusion 2002;42:37–43.
transfusion reactions. Am J Clin Pathol 1980;74:94–97. 78. Campbell SA, Shirey RS, King KE, et al: An acute hemolytic
55. Taswell HF, Pineda AA, Moore SB: Hemolytic transfusion transfusion reaction due to anti-IH in a patient with sickle cell
reactions: Frequency and clinical and laboratory aspects. In disease. Transfusion 2000;40:828–831.
Bell CA (ed): A Seminar on Immune-mediated Cell De- 79. Win N, Doughty H, Telfer P, et al: Hyperhemolytic transfusion
struction. Washington, DC: American Association of Blood reaction in sickle cell disease. Transfusion 2001;41:323–328.
Banks, 1981:71. 80. Chaplin H, Cassell M: The occasional fallibility of in vitro
56. Vamvakas EC, Pineda AA, Reisner R, et al: The differentiation compatibility tests. Transfusion 1962;2:375.
of delayed hemolytic and delayed serologic transfusion reac- 81. Reed W, Walker P, Haddix T, et al: Acute anemic events in
tions: Incidence and predictors of hemolysis. Transfusion sickle cell disease. Transfusion 2000;40:267–273.
1995;35:26–32. 82. Chaplin H, Zarkowsky HS: Combined sickle cell disease and
57. Pineda AA, Vamvakas EC, Gorden LD, et al: Trends in the autoimmune hemolytic anemia. Arch Intern Med
incidence of delayed hemolytic and delayed serologic trans- 1981;141:1091–1093.
fusion reactions. Transfusion 1999;39:1097–1103. 83. Sosler SD, Perkins JT, Saporito C, et al: Severe autoimmune
58. Fudenberg H, Allen FH: Transfusion reactions in the absence of hemolytic anemia induced by transfusion in two alloimmu-
demonstrable incompatibility. N Eng J Med 1957;256:1180. nized patients with sickle cell disease. Transfusion 1989;
59. Croucher BEE, Crookston MC, Crookston JH: Delayed 29:49S.
haemolytic transfusion reactions simulating auto-immune 84. Tolano J, Hillary CA, Gottschall JL, et al: Delayed hemolytic
haemolytic anemia. Vox Sang 1967;12:32. transfusion reaction/hyperhemolysis syndrome in children
60. Ness PM, Shirey RS, Thoman SK, et al: The differentiation of with sickle cell disease. Pediatrics 2003;111:661–665.
delayed serologic and delayed hemolytic transfusion reac- 85. Sirchia G, Morelati F, Rebulla P: The sickle cell hemolytic
tions: Incidence, long-term serologic findings, and clinical transfusion reaction syndrome [letter]. Transfusion 1997;
significance. Transfusion 1990;30:688–693. 37:1098–1099.
61. Pinkerton PH, Coovadia AS, Goldstein J: Frequency of 86. Grainger JD, Makar Y, McManus A, et al: Refractory hyper-
delayed hemolytic transfusion reactions following antibody haemolysis in a patient with beta-thalassaemia major. Transfus
screening and immediate-spin crossmatching. Transfusion Med 2001;11:55–57.
1992;32:814–817. 87. Petz LD: Blood transfusion in hemolytic anemias. Immuno-
62. Salama A, Mueller-Eckhardt C: Delayed hemolytic transfusion hematology 1999;15:15–23.
reactions: Evidence for complement activation involving allo- 88. Petz LD: The expanding boundaries of transfusion medicine.
geneic and autologous red cells. Transfusion 1984;24:188–193. In Nance SJ (ed): Clinical and Basic Science Aspects of
63. Solanki D, McCurdy PR: Delayed hemolytic transfusion reac- Immunohematology. Arlington, VA: American Association of
tions: An often-missed entity. JAMA 1978;239:729–731. Blood Banks, 1991:73–113.
64. Diamond WJ, Brown FL Jr, Bitterman P, et al: Delayed 89. Petz LD, Garratty G: Acquired immune hemolytic anemias,
hemolytic transfusion reaction presenting as sickle-cell crisis. 1st ed. New York: Churchill Livingstone, 1980.
Ann Intern Med 1980;93:231–234. 90. Adamson JW: The erythropoietin/hematocrit relationship in
65. Millner PF, Squires JE, Larison PJ, et al: Posttransfusion crises normal and polycythemic man: Implications of marrow regu-
in sickle cell anemia: Role of delayed hemolytic transfusion lation. Blood 1968;32:597–609.
reactions to transfusion. South Med J 1985;78:1462–1469. 91. Beutler E: Production and destruction of erythrocytes. In
66. Millner PF, Squires JE: Post-transfusion crises in sickle cell Beutler E, Lichtman MA, Coller BS, Kipps TJ, Seligsohn U
anemia. Prog Clin Biol Res 1987;240:351–359. (eds): Williams Hematology, 6th ed. New York: McGraw-Hill,
67. Rao KR, Patel AR: Delayed hemolytic transfusion reactions in 2001:355–368.
sickle cell anemia. South Med J 1989;82:1034–1036. 92. Hillman RS, Finch CA: Erythropoiesis: Normal and abnormal.
68. Petz LD, Shulman IA: The sickle cell-hemolytic transfusion Seminars Hematol 1967;4:327–336.
reaction syndrome. Proceedings of the International Society of 93. Mann JR, Cotter KP, Walker R, et al: Anaemic crisis in sickle
Blood Transfusion/American Association of Blood Banks cell disease. J Clin Pathol 1975;28:341–344.
Joint Congress. Los Angeles, 1990:164. 94. Ballas SK, Gay RN: The sickle cell hemolytic transfusion reac-
69. Orlina AR, Sosler SD, Koshy M: Problems of chronic transfu- tion syndrome. Transfusion 1997;37:1099–1100.
sion in sickle cell disease. J Clin Apheresis 1991;6:234–240. 95. Diggs LW: The crisis in sickle cell anemia: Hematological
70. Cummins D, Webb G, Shah N, Davies SC: Delayed hemolytic studies. Am J Clin Path 1956;26:1109–1118.
transfusion reactions in patients with sickle cell disease. 96. Diggs LW: Sickle crises. Am J Clin Path 1965;44:1–19.
Postgrad Med J 1991;67:689–691. 97. Naumann HN, Diggs LW, Barreras L, et al: Plasma hemoglo-
71. Lankiewicz MW, Shirey RS, Ness PM, Charache S: Delayed bin and hemoglobin fractions in sickle cell crisis. Am J Clin
hemolytic transfusion reactions (DHTR) in sickle cell disease Pathol 1971;56:137–147.
570 Immune Hemolytic Anemias

98. Telen MJ: Principles and problems of transfusion in sickle cell 122. Mummert TB, Tourault MA: Review of transfusion related
disease. Semin Hematol 2001;38:315–323. fatalities: Many preventable. Hosp Technol Scanner 1993;11:1–3.
99. Beauregard P, Blajchman MA: Hemolytic and pseudo- 123. Linden JV, Paul B, Dressler KP: A report of 104 transfusion
hemolytic transfusion reactions: An overview of the hemolytic errors in New York State. Transfusion 1992;32:601–606.
transfusion reactions and the clinical conditions that mimic 124. Linden JV, Kaplan HS: Transfusion errors: Causes and effects.
them. Transfus Med Rev 1994;8:184–199. Trans Med Rev 1994;VIII:169–183.
100. Crookston JH: Transfusion reactions. Am J Med Technol 1968; 125. Williamson L, Cohen H, Love E, et al: The serious hazards of
334:579–588. transfusion (SHOT) initiative: The UK approach to haemovig-
101. Davenport RD, Strieter RM, Standiford TJ, et al: Interleukin-8 ilance. Vox Sang 2000;78:291–295.
production in red blood cell incompatibility. Blood 126. Ibojie J, Urbaniak SJ: Comparing near misses with actual mis-
1990;76:2439–2442. transfusion events: A more accurate relection of transfusion
102. Davenport RD, Strieter RM, Kunkel SL: Red cell ABO incom- errors. Br J Haematol 2000;108:458–460.
patibility and production of tumour necrosis factor-alpha. Br J 127. Callum JL, Kaplan HS, Merkley LL, et al: Reporting of near-
Haematol 1991;78:540–544. miss events for transfusion medicine: Improving transfusion
103. Butler J, Parker D, Pillai R, et al: Systemic release of neutrophil safety. Transfusion 2001;41:1204–1211.
elastase and tumour necrosis factor alpha following ABO 128. Taswell HF, Galbreath JL, Harmsen WS: Errors in transfusion
incompatible blood transfusion. Br J Haematol 1991;79:525–526. medicine. Arch Pathol Lab Med 1994;118:405–410.
104. Davenport RD, Burdick M, Strieter RM, et al: Monocyte 129. Baele PL, DeBruyere M, Deneys V, et al: Bedside transfusion
chemoattractant protein production in red cell incompatibil- errors. Vox Sang 1994;66:117–121.
ity. Transfusion 1994;34:16–19. 130. Shulman IA, Lohr K, Derdiarian AK, et al: Monitoring trans-
105. Davenport RD, Burdick MD, Strieter RM, et al: In vitro pro- fusionist practices: A strategy for improving transfusion
duction of interleukin-1 receptor antagonist in IgG-mediated safety. Transfusion 1994;34:11–15.
red cell incompatibility. Transfusion 1994;34:297–303. 131. Shulman IA, Jones FS: Practical aspects of the self-assessment
106. Capon SM, Goldfinger D: Acute hemolytic transfusion reac- of the issue and administration of blood. Am J Clin Pathol
tion, a paradigm of the systemic inflammatory response: New 1997;104(S1):S17–S22.
insights into pathophysiology and treatment. Transfusion 132. Whitsett CF, Robichaux MG: Assessment of blood administra-
1995;35:513–520. tion procedures: Problems identified by direct observation
107. Rabiner SF, Friedman LH: The role of intravascular haemoly- and administrative incident reporting. Transfusion 2001;
sis and the reticuloendothelial system in the production of a 41:581–586.
hypercoagulable state. Br J Haematol 1968;14:105–118. 133. Shulman IA, Kent D: Unit placement errors: A potential risk
108. Birndorf NL, Lopas H, Robboy SJ: Disseminated intravascular factor for ABO and Rh incompatible blood transfusions. Lab
coagulation and renal failure: Production in the monkey with Med 1991;22:194–196.
autologous red blood cell stroma. Lab Invest 1971;25:314–319. 134. Renner SW, Howanitz PJ, Bachner P: Wristband identification
109. Poskitt TR, Fortwengler HP Jr, Lunskis BJ: Activation of the error reporting in 712 hospitals. Arch Pathol Lab Med
alternate complement pathway by autologus red cell stroma. 1993;117:573–577.
J Exp Med 1973;138:715–722. 135. Dale JC, Renner SW: Wristband errors in small hospitals. Lab
110. Schmidt PJ, Holland PV: Pathogenesis of the acute renal Med 1997;28:203–207.
failure associated with incompatible transfusion. Lancet 136. Lau FY, Wong R, Chui CH, et al: Improvement in transfusion
1967;2:1169–1172. safety using a specially designed transfusion wristband. Trans
111. Feinstein D: Diagnosis and management of disseminated Med 2000;10:121–124.
intravascular coagulation: The role of heparin therapy. Blood 137. Wenz B, Burns ER: Improvement in transfusion safety using a
1982;60:284–287. new blood unit and patient identification system as part of
112. Goldfinger D: Acute hemolytic transfusion reactions—a fresh safe transfusion practice. Transfusion 1991;31:401–403.
look at pathogenesis and considerations regarding therapy. 138. Mercuriali F, Inghilleri G, Colotti MT, et al: Bedside transfu-
Transfusion 1977;17:85–98. sion errors: analysis of 2 years’ use of a system to monitor and
113. Rock RC, Bove JR, Nemerson Y: Heparin treatment of prevent transfusion. Vox Sang 1996;70:16–20.
intravascular coagulation accompanying hemolytic transfu- 139. AuBuchon JP, Littenberg B: A cost-effectiveness analysis of the
sion reactions. Transfusion 1969;9:57–61. use of a mechanical barrier system to reduce the risk of mis-
113a. Linden JV, Wagnerm K, Voytovich AE, Sheehan J: Transfusion transfusion. Transfusion 1996;36:222–226.
errors in New York State: An analysis of 10 years experience. 140. Jensen NJ, Crosson JT: An automated system for bedside
Transfusion 2000;40:1207–1213. verification of the match between patient identification and
114. Buchholz DH, Bove JR: Unusual response to ABO incompati- blood unit identification. Transfusion 1996;36:216–221.
ble blood transfusion. Transfusion 1975;15:577–582. 141. Langeberg AF, Berg M, Novak SC, et al: Evaluation of the
115. Rydberg L, Breimer ME, Samuelsson BE: Antibody response Immucor I-Trac system for positive patient, blood sample and
in an ABO-incompatible blood transfusion. Transfusion blood unit identification [abstract]. Transfusion 1999;39:25S.
1988;28:483–438. 142. Lumadue JA, Boyd JS, Ness PM: Adherence to a strict speci-
116. Bacon JM, Young IF: ABO incompatible blood transfusion. men-labeling policy decreases the incidence of erroneous
Pathology 1989;21:181–184. blood grouping of blood bank specimens. Transfusion
117. Garratty G, Arndt P, Co A, et al: Fatal hemolytic transfusion 1997;37:1169–1172.
reaction resulting from ABO mistyping of a patient with 143. Snyder EL, Spivack M, Novodoff D, et al: Hemolytic anti-
acquired B antigen detectable only by some monoclonal anti- hr”(e) detectable solely by an automated Polybrene technique.
B reagents. Transfusion 1996;36:351–357. Transfusion 1978;18:79–83.
118. Schmidt PJ: The mortality from incompatible transfusion. In 144. Garratty G, Vengelen-Tyler V, Postoway N, et al: Hemolytic
Sandler GS, Nusbracher J, Schanfield MS (eds): Immuno- transfusion reaction (HTR) associated with antibodies not
biology of the erythrocyte. New York: Liss, 1980:251–261. detectable by routine procedures [abstract]. Transfusion
119. Myhre BA: Fatalities from blood transfusion. JAMA 1982;22:429.
1980;244:1333–1335. 145. Garratty G, Arndt P, Postoway N, et al: Severe hemolytic trans-
120. Honig CL, Bove JR: Transfusion-associated fatalities: Review fusion reactions associated with antibodies not detectable by
of Bureau of Biologics reports 1976–1978. Transfusion routine methods [abstract]. Transfusion 1986;36:23S.
1980;20:653–661. 146. Maynard BA, Smith DS, Farrar RP, et al: Anti-Jka, -C, -E in a
121. Sazama K: Reports of 355 transfusion-associated deaths: 1976 single patient, initially demonstrable only by the manual
through 1985. Transfusion 1990;30:583–590. hexadimethrine bromide (Polybrene) test, with incompatibili-
Hemolytic Transfusion Reactions 571

ties confirmed by 51Cr-labeled red cell studies. Transfusion 169. Aubert EF, Boorman KE, Dodd BE, Loutit JF: The universal
1988;28:302–306. donor with high titre iso-agglutinins. Br Med J 1942;659–664.
147. Devenish A, Kay LA: Hemolytic transfusion reactions due to 170. Inwood MJ, Zuliani B: Anti-A hemolytic transfusion with
anti-e+f detectable only by nonstandard serologic techniques. packed O cells. Annals Int Med 1978;89:515–516.
Immunohematology 1994;10:120–123. 171. Boothe G, Brecher ME, Root M, Robinson J, Haley R: Acute
148. Lin CK, Wong KT, Mak KH, et al: Hemolytic transfusion reac- hemolysis due to passively transfused high-titer anti-B
tion due to Rh antibodies detectable only by manual causing spontaneous in vitro agglutination. Immunohema-
Polybrene and polyethylene glycol methods. Am J Clin Pathol tology 1995;11:43–45.
1995;104:660–662. 172. Crosby WH, Akeroyd JH: Some immunohematologic results
149. Lown J, Willis J: Monocyte monolayer assay (MMA) reactivity of large transfusions of group O blood in recipients of other
of alloantibodies reacting by the manual polybrene technique blood groups. Blood 1954;IX:103–116.
but not by an antiglobulin test. Transf Med 1995;5:281–284. 173. Barnes A Jr: Status of the use of universal donor blood trans-
150. Stewart JW, Mollison PL: Rapid destruction of apparently fusion. CRC Crit Rev Clin Lab Sci 1973;4:147–160.
compatible red cells. Br Med J 1959;i:1274. 174. Schwab CW, Shayne JP, Turner J: Immediate trauma resuscita-
151. Kissmeyer-Nielsen F, Jensen KB, Ersbak J: Severe haemolytic tion with type O uncrossmatched blood: A two-year prospec-
transfusion reactions caused by apparently compatible red tive experience. J Trauma 1986;26:897–902.
cells. Br J Haematol 1961;7:36. 175. Standards for blood banks and transfusion services, 22nd ed.
152. van Loghem JJ, van der Hart M, Moes M, et al: Increased red Bethesda, MD: American Association of Blood Banks, 2003.
cell destruction in the absence of demonstrable antibodies in 176. Jennings ER, Hindmarsh C: The significance of the minor
vitro. Transfusion 1965;5:525. crossmatch. Am J Clin Pathol 1958;30:302–313.
153. Davey RJ, Gustafson M, Holland PV: Accelerated immune red 177. Mohn JR, Lambert RM, Bowman HS, Brason FW:
cell destruction in the absence of serologically detectable Experimental transfusion of donor plasma containing blood-
alloantibodies. Transfusion 1980;20:348–353. group antibodies into incompatible normal human recipients.
154. Baldwin ML, Barrasso C, Ness PM, et al: A clinically I. Absence of destruction of red-cell mass with anti-Rh, anti-
significant erythrocyte antibody detectable only by 51Cr sur- Kell and anti-M. Br J Haematol 1961;17:112.
vival studies. Transfusion 1983;23:40–44. 178. Bowman HS, Brason FW, Mohn JF, Lambert RM: Experi-
155. Halima D, Brunt D, Postoway N, et al: Hemolytic transfusion mental transfusion of donor plasma containing blood-
reactions (HTR) due to a probable anti-C, not detectable by group antibodies into incompatible normal human recipients.
multiple technics [abstract]. Transfusion 1982;22:405. II. Induction of isoimmune haemolytic anaemia by a transfu-
156. Harrison CR, Hayes TC, Trow LL, et al: Intravascular sion of plasma containing exceptional anti-CD antibodies. Br
hemolytic transfusion reaction without detectable antibodies: J Haematol 1961;7:130.
A case report and review of the literature. Vox Sang 179. Goldfinger D, Kleinman S, Connelly M, Chaux A, Sacks HJ:
1986;51:96–101. Acute hemolytic transfusion reaction (HTR) with dissemi-
157. van der Hart M, Szaloky A, van den Berg-Loonen EM, et al: nated intravascular coagulation (DIC) and acute renal failure
Présence d’ antigénes HL-A sur les hématies d’un donneur (ARF) due to transfusion of plasma containing Rh antibodies
normal. Nouv Revue Fr Hémat 1974;14:555–563. [abstract]. Transfusion 1979;19:639.
158. Nordhagen R, Aas M: Association between HLA and red cell 180. Scaradavou A, Woo B, Woloski BMR, et al: Intravenous anti-D
antigens. VII. Survival studies of incompatible red blood cells treatment of ITP: Experience in 272 patients. Blood 1997;
in a patient with HLA-associated haemagglutinins. Vox Sang 89:2689–2700.
1978;35:319–323. 181. Zettner A, Bove JR: Hemolytic transfusion reaction due to
159. Panzer S, Mueller-Eckhardt G, Salama A, et al: The clinical interdonor incompatibility. Transfusion 1963;3:48–51.
significance of HLA antigens on red cells. Transfusion 182. Franciosi RA, Awer E, Santana M: Interdonor incompatibility
1984;24:486–489. resulting in anuria. Transfusion 1967;7:297–298.
160. Panzer S, Mayr WR, Graninger W, et al: Haemolytic transfu- 183. Abbott D, Hussain S: Intravascular coagulation due to inter-
sion reactions due to HLA antibodies. Lancet 1987;1:474–478. donor incompatibility. Can Med Assoc J 1970;103:752–753.
161. Mollison PL, Engelfriet CP, Contreras M: Blood transfusion in 184. West NC, Jenkins JA, Johnston BR, Modi N: Interdonor
clinical medicine, 9th ed. Oxford: Blackwell Scientific, incompatibility due to anti-Kell antibody undetectable by
1993:461–462. automated antibody screening. Vox Sang 1986;50:174–176.
162. Weitekamp LA, Johnson ST, Larson LB, et al: Severe, sympto- 185. Morse EE: Interdonor incompatibility as a cause of reaction
matic haemolytic transfusion reactions secondary to strong during granulocyte transfusion. Vox Sang 1978;35:215–218.
HLA antibody directed toward A2 public and private epi- 186. Reiner AP, Sayers MH: Hemolytic transfusion reaction due to
topes [abstract]. Transfusion 1993;33:55S. interdonor Kell incompatibility. Arch Pathol Lab Med
163. Benson K, Agosti SJ, Latoni-Benedetti GE, Leparc GF: Acute 1990;114:862–864.
and delayed hemolytic transfusion reactions secondary to 187. Gover PA, Morton JR: Transfusion reaction due to anti-Fya in
HLA alloimmunization. Transfusion 2003;43:753–757. donor blood. Clin Lab Haemat 1990;12:233–236.
164. Garratty G: Basic mechanisms of in vivo cell destruction. In 188. Naczek CA, Mikulski DA, Kavemeier K, et al: Safety of trans-
Bell CA (ed): A Seminar on Immune-mediated Cell fusing red blood cells from donors with red cell alloantibodies
Destruction. Washington, DC: American Association of Blood [abstract]. Transfusion 1992;32:25S.
Banks, 1981:1–28. 189. Combs MR, Bennett DH, Telen MJ: Large-scale use of red
165. Gilsanz F, de la Serna FJ, Molt L, et al: Haemolytic anaemia blood cell units containing alloantibodies. Immuhematology
associated to chronic large granular lymphocytic (LGG) 2000;16:120–123.
leukemia of NK-cells: Cytotoxicity of NK cells against RBC 190. Lundberg WB, McGinniss MH: Hemolytic transfusion reac-
[abstract]. Blood 1994;84:183a. tion due to anti-A1. Transfusion 1975;15:1–9.
166. Garratty G: Problems associated with passively transfused 191. Zoes C, Dube VE, Miller HJ, Vye MV: Anti-A1 in the plasma
blood group alloantibodies. Am J Clin Pathol 1998;109:769–777. of platelet concentrates causing a hemolytic reaction.
167. Rieben R, Buchs JP, Flückiger E, Nydegger UE: Antibodies to Transfusion 1977;17:29–32.
histo-blood group substances in A and B: Agglutination titers, 192. Thomas L, Tuscany FL, Wilner GD: Anti-A antibody trans-
Ig class, and IgG subclasses in healthy persons of difference fused with non-group-specific platelets. Arch Pathol Lab Med
age categories. Transfusion 1991;31:607–615. 1981;105:52–53.
168. Auf Der Maur C, Hodel M, Nydegger UE, Rieben R: Age 193. Siber GR, Ambrosino DM, Gorgone BC: Blood-group-A-like
dependency of ABO histo-blood group antibodies: substance in a preparation of pneumococcal vaccine. Annals
Reexamination of an old dogma. Transfusion 1993;33:915–918. Int Med 1982;96:580–586.
572 Immune Hemolytic Anemias

194. McLeod BC, Sassetti RJ, Weens JH, Vaithianathan T: Haemolytic 204. Larsson LG, Welsh VJ, Ladd DJ: Acute intravascular hemo-
transfusion reaction due to ABO incompatible plasma in a lysis secondary to out-of-group platelet transfusion. Trans-
platelet concentrate. Scand J Haematol 1982;28:193–196. fusion 2000;40:902–906.
195. Conway LT, Scott EP: Acute hemolytic transfusion reaction 205. Rosati LA, Barnes B, Oberman HA, Penner JA: Hemolytic
due to ABO incompatible plasma in a plateletapheresis con- anemia due to anti-A in concentrated antihemophilic factor
centrate. Transfusion 1984;34:413–414. preparations. Transfusion 1970;10:139–141.
196. Pierce RN, Reich LM, Mayer K: Hemolysis following platelet 206. Seeler RA: Hemolysis due to anti-A and anti-B in factor VIII
transfusions from ABO-incompatible donors. Transfusion preparations. Arch Int Med 1972;130:101–103.
1985;25:60–62. 207. Ragni MV, Spero JA, Larson P, Ramsey G, Lewis JH: Immune
197. Ferguson DL: Acute intravascular hemolysis after a platelet hemolysis by isohemagglutinins in factor IX concentrates.
transfusion. CMAJ 1988;138:523–524. Transfusion 1987;27:292–293.
198. Reis MD, Coovadia AS: Transfusion of ABO-incompatible 208. Orringer EP, Koury MJ, Blatt PM, Roberts HR: Hemolysis
platelets causing severe haemolytic reaction. Clin Lab Hemat caused by factor VIII concentrates. Arch Intern Med
1989;11:237–240. 1976;136:1018–1020.
199. Murphy MF, Hook S, Waters AH, et al: Acute haemolysis after 209. Ashenhurst JB, Langehenning PL, Seeler RA, Telfer MC:
ABO-incompatible platelet transfusions. Lancet 1990;335: Hemolytic anemia due to anti-B in antihemophiliac factor
974–975. concentrates. J Pediatr 1976;88:257–258.
200. Take H, Jinbo T, Tamura J, et al: Fatal intravascular hemolysis 210. Seeler RA, Telischi M, Langehenning PL, Ashenhurst JB:
induced by platelet concentrate. Am J Hematol 1994;46:251–252. Comparison of anti-A and anti-B titers in factor VIII and IX
201. Shanwell A, Ringdén O, Wiechel B, Rumin S, Åkerblom O: A concentrates. J Pediatr 1976;89:87–89.
study of the effect of ABO incompatible plasma in platelet 211. Buchanan GR, Holtkamp CA, Johnson A: Reduced serum
concentrates transfused to bone marrow transplant recipients. haptoglobin values in hemophiliacs receiving monclonally
Vox Sang 1991;60:23–27. purified factor VIII concentrates. Am J Hematol 1990;
202. Chipping PM, Lloyd E, Goldman JM: Haemolysis after gran- 33:234–237.
ulocyte transfusions. Br Med J 1980;281:1529–1530. 212. Masouredis SP, Chaplin H Jr: Transfusion management
203. Mair B, Benson K: Evaluation of changes in hemoglobin levels of autoimmune hemolytic anemia. In Chaplin H Jr (ed):
associated with ABO-incompatible plasma in apheresis Immune Hemoytic Anemias. New York: Churchill Livingstone,
platelets. Transfusion 1998;38:51–55. 1985:177.
Index
Note: Page numbers followed by f indicate an illustration; page numbers followed by t indicate a table.

A antigen. See also ABO blood group. Acanthocytes, 54–55 Acquired immunodeficiency syndrome. See
adsorption of, in bystander immune Ackroyd hapten hypothesis, for DIIHA, Human immunodeficiency virus
hemolysis, 362–363, 363f 265–266 infection.
autoantibodies to, 238, 251–252 Acquired autoimmune hemolytic Acrocyanosis, in cold agglutinin syndrome,
transfer of, in group O blood transfusion, anemia(s), 319–374 72, 167
566 versus delayed transfusion reactions, Actin, autoantibodies to, 244
ABO blood group 356–357 Activation unit, in complement activation,
antibodies to familial, 351–352, 352t 134, 136, 137f
HDFN in, 535–536 laboratory tests for, in pediatric patients, Acute transient autoimmune hemolytic
in solid organ transplantation, 501t, 342–343, 342t anemia, 74
502 negative DAT in, 319–334 Acute tubular necrosis, in hemoglobinuria,
autoantibodies to, 238–239, 358 antibody concentration versus 49
determination of, in positive DAT, 215 hemolysis rate in, 320–321, Adenylate kinase, erythrocyte, 50
in hematopoietic cell transplantation 320f, 320t, 321f Adsorption procedures
glycosyltransferase activities after, case reports of, 323–325 for alloantibody detection
471–472, 472f, 473t course of, 334 in AIHA, 542
graft-versus-host disease and, 481–483, diagnosis of, 332–333, 332t, 333t allogeneic, 384–385, 390, 390t
482f functional cellular assays and, autologous cell storage for, 384
major incompatibility, 473–478 333–334 overview of, 380, 382
delayed hemolytic reactions in, idiopathic, 323–324 posttransfusion timing of, 383–384, 384t
473–474 Ig measurement in, 321–329, 322t, 323t, in recently transfused patients,
differential diagnosis of, 493–494 325t, 328f 382–383, 383t
hemolysis incidence in, 495 IgA with, 330–331 warm autoadsorption technique for,
isohemagglutinin persistence after, IgM with, 330–332 382–383
474–476, 474f, 475f incidence of, 319–320 cold, 390, 390t
management of, 478–481, 479t, 480f, low-affinity Ig with, 329–330, 330f African trypanosomiasis, AIHA in, 109
481f in pregnancy, 347–349, 348f, 348t Age factors
pure red cell aplasia in, 476–477, recurrent, 323–324 in positive DAT results, 206
476f–478f sensitivity of, 321 in postsplenectomy infections, 413t
minor incompatibility, 460–468, 460f, in sickle cell disease, 325–327, 325t Agglutination. See also Isohemagglutinins.
460t treatment of, 334 in cold agglutinin syndrome, 72–73, 168,
bystander hemolysis in, 463–464 in pediatric patients, 341–344 168f
differential diagnosis of, 493 clinical findings in, 341–342, 341t, 342f, in direct globulin test, 202, 202f
graft-versus-host disease 342t early description of, 4–5, 4f, 6f, 7f
prophylaxis in, 467–468 laboratory findings in, 342–343, 342t grading system for, 206, 207t
isohemagglutinin persistence after, prognosis for, 344 mechanisms of, 161
473 treatment of, 343–344 spontaneous, in positive DAT, 214–217
management of, 468–471, 470t very young, 343–344 in warm antibody AIHA, 66, 168
massive hemolysis in, 463–467, in pregnancy. See Pregnancy, AIHA in. Agnogenic myeloid metaplasia, AIHA in,
464f–466f after transfusion, 335–340 113
passenger lymphocyte syndrome in, animal studies of, 335–336 AIDS. See Human immunodeficiency virus
467, 469–470, 470t, 493 autoantibody source in, 339–340 infection.
serologic findings in, 461–463, 461t case reports of, 336–337 AIHA. See Autoimmune hemolytic
transfusion requirements in, 463, evidence of, 335, 335t anemia(s).
464t immunologic data on, 338–339 Alloantibody(ies)
in transfusion reactions, incompatibility multiple, 337–338 autoantibodies mimicking, 242
of, 558–559, 566 Acquired hemolytic anemias, 52t, 53t. in bystander immune hemolysis. See
Abortion, in AIHA, 346 See also specific types, eg, Acquired Bystander immune hemolysis.
Abrami, P., 13–14 autoimmune hemolytic anemia(s). cross-reacting, bystander immune
Absolute reticulocyte count, 36 versus congenital hemolytic anemia, 5, 7, hemolysis and, 364
direct determination of, 39–40 13–14, 15f, 16f detection of, in presence of
in red blood cell life span calculation, 39, definition of, 33 autoantibodies, 379–385,
40f early descriptions of, 2 381t–384t
574 Index

Alloantibody(ies) (continued) Antibody(ies) (continued) Asplenia. See Splenectomy.


formation of, after hematopoietic cell specific adsorption of, for HDFN, 534 Autoadsorption
transplantation, 488–490, 488t specificity of. See also Specificities, of cold, 390, 390t
mimicking autoantibodies, 239–242, 241t autoantibodies. warm
multiple, after solid organ determination of, 224–226, 225t, 226t autologous cell storage for, 384
transplantation, 504–505, 505f synthesis of, in warm antibody AIHA overview of, 380, 382
passive transfusion of, 566–567 corticosteroid effects on, 404–405, 405t posttransfusion waiting time for,
of recipient origin, after solid organ splenectomy effects on, 410 383–384, 384t
transplantation, 505–506 undetectable by routine procedures, technique for, 382–383, 383t
risks from, in transfusions, 378 563–567 Autoagglutination
test frequency for, in multiple but detected by special procedures, in cold agglutinin syndrome, 72–73, 168,
transfusions, 387–388 563–564 168f
in transfusion reactions, 336–337 later appearance of, 563 in warm antibody AIHA, 66, 168, 168f
in transplantation, 459–460 Antibody-dependent cell-mediated Autoantibodies. See also specific
in warm antibody AIHA, 379, 379t cytotoxicity autoantibodies.
Alloantibody-induced hemolytic anemia, in DAT-negative AIHA, 334 in DIIHA, 287–290, 287t, 288t, 290f
differential diagnosis of, 168 in HDFN, 527 discovery of, 12–13, 14f
Allogeneic adsorption test, 384–385, 390, in red blood cell destruction, 158 drug-induced, 272
390t Anticardiolipin antibodies, 245 low-affinity, detection of, 213
Alloimmune thrombocytopenia in AIHA, 94, 96, 445 mimicking alloantibodies, 242
after hematopoietic cell transplantation, in warm antibody AIHA, 65 risks from, in transfusions, 378
491–492 Anticoagulants, for warm antibody AIHA, specificities of. See Specificities, of
after solid organ transplantation, 506 66 autoantibodies.
Alloimmunity, definition of, 459–460 Antigen(s). See also specific antigens. after transfusions, 335–340, 335t
Alloimmunization antibody complex with. See Immune in transplantation, 459–460
bystander immune hemolysis and, 364 complex(es). Autoimmune disease. See also specific
in solid organ transplantation, 502, Antiglobulin serum (AGS), 202. See also diseases.
505–506 Anti-human globulin (AHG). after hematopoietic cell transplantation,
in transfusions, incidence of, 541–543 adsorption of, for phenotyping, 217 490
Alpha interferon. See Interferon-α. anti-complement, standardization of, Autoimmune hemolytic anemia(s)
Amniocentesis, for HDFN detection, 211–212 acquired. See Acquired autoimmune
521–524, 523f for complement detection, 204 hemolytic anemia(s).
Amoxicillin, DIIHA from, 286 contamination of, 204 acute, transfusions for, 376
Amoxicillin/clavulanate, DIIHA from, 282 dilution of, for titration, 172, 172t, 173f, alloimmunization in, 542
Ampicillin, DIIHA from, 286 174, 174f, 175t, 176t antibody characterization in, 177–178
Ampicillin/sulbactam, DIIHA from, 282 for IgG detection, 204 antibody specificity in
Andral, Gabriel, 2, 3f IgG subclass, 213–214 membrane components, 243–244
Anemia, with hemolytic component, inappropriate antibodies in, 204 in stored red blood cells, 243, 253–254
definition of, 33 quality control of, 212 in younger red blood cells, 244
Angina, in severe anemia, 377 special, for DAT-negative AIHA, 332 chronic stable, transfusions for, 377
Angioimmunoblastic lymphadenopathy test sensitivity and, 321 classification of, 61–62, 62t, 201
with dysproteinemia Antiglobulin (Coombs) test. See also Direct cold agglutinin syndrome as. See Cold
AIHA in, 112–113 antiglobulin test; Indirect agglutinin syndrome.
autoantibodies in, 245 antiglobulin test. concept of, 23–24
Anisocytosis in AIHA, in ovarian tumors, 85t definition of, 1
in cold agglutinin syndrome, 73 capillary method, 214 differential diagnosis of. See under
in sickle cell disease, 55f development of, 20–21, 23, 23f Differential diagnosis.
in warm antibody AIHA, 66 enzyme-linked, in warm antibody AIHA, direct antiglobulin rest results in,
Anti-dl (deleted) autoantibodies, 232, 232t, 179–180 170–174, 171t, 172t, 173f, 174f,
233, 235, 235t principles of, 202–203, 202f, 202t 175t, 176t
Anti-human globulin (AHG), 202, 202f. See reagents for, 203 versus drug-induced immune hemolytic
also Antiglobulin serum (AGS). sedimentation method, 214 anemia, 168
monospecific, 170–172, 171t, 203 tube method, 213–214 first description of, 11
polyspecific, 170–172, 171t, 203 uses of, 202, 202t fulminant
Anti-nl (normal) autoantibodies, 232, 232t, Antilymphocyte globulin, transfusion of, plasma exchange for, 430
233, 235, 235t, 385 hemolysis after, 496 transfusions for, 377
Anti-pdl (partially deleted) autoantibodies, Antiphospholipid antibodies and after hematopoietic cell transplantation,
232, 232t, 233, 235, 235t antiphospholipid syndrome, 245 483–487, 484t–486t, 487f
Antibiotics, for postsplenectomy infections, in AIHA, 94, 96 differential diagnosis of, 494–496, 497t
413, 414, 414t in warm antibody AIHA, 65 versus immune hemolytic anemia from
Antibody(ies). See also Autoantibodies; Aplastic anemia, hematopoietic cell multiple alloantibodies,
specific antibodies. transplantation for, AIHA after, 504–505, 505f
antigen complex with. See Immune 487 mixed warm and cold. See Mixed
complex(es). Apoptosis, defects of, in autoimmune autoimmune hemolytic anemia.
avidity of, in warm antibody AIHA, lymphoproliferative syndrome, paroxysmal cold hemoglobinuria as. See
corticosteroid effects on, 405 112 Paroxysmal cold
characterization of, screen for, 219–220, Apronalide, DIIHA from, 268 hemoglobinuria (PCH).
221t APT1 gene mutations, in autoimmune rapidly progressive, transfusions for, 376,
low-affinity, false-negative DAT in, 204 lymphoproliferative syndrome, 377, 377t
passive transfer of, in hematopoietic cell 112 refractory, rituximab for, 420–422, 420f
transplantation, 494–496, 497t Armed macrophages, in red blood cell screening tests in, 177–178
red blood cell. See Red blood cell destruction, 160–161, 160f secondary. See Secondary autoimmune
antibodies. Ashby, Winifred, 18–19, 19f hemolytic anemia(s).
Index 575

Autoimmune hemolytic anemia(s) Blood film, 53–58, 54f–57f Campath-1H, for AIHA, in chronic
(continued) in warm antibody AIHA, 66 lymphocytic leukemia, 444
in sickle cell disease, 553–555, 554f Blood groups. See also specific blood groups, Canale-Smith syndrome (autoimmune
after solid organ transplantation, 506 eg, ABO blood group. lymphoproliferative syndrome),
stable, transfusions for, 376–377 determination of, in DAT-positive AIHA in, 112, 112t
transfusions for. See Transfusion(s), for patients, 214–215 Cancer
AIHA. Blood transfusions. See Transfusion(s). AIHA in, 97, 97t, 98t
warm antibody. See Warm antibody Bone, corticosteroid effects on cold agglutinin syndrome in, 113t, 114
autoimmune hemolytic anemia. avascular necrosis, 406–407 hematologic. See Leukemia; Lymphoma.
without serum autoantibody, 387 growth abnormalities, 408 from immunosuppressive therapy, 418
Autoimmune lymphoproliferative loss (osteoporosis), 406 ovarian, AIHA in, 83–84, 85t
syndrome, 112, 112t Bone marrow Candidiasis, mucocutaneous, AIHA in, 111,
Autoimmune thrombocytopenia, after in cold agglutinin syndrome, 73 111t
hematopoietic cell transplantation, in paroxysmal cold hemoglobinuria, 76 Carbimazole, DIIHA from, 268
491 in warm antibody AIHA, 69 Carbodiimide, for DIIHA testing, 307
Autoimmunity, definition of, 459–460 Bone marrow transplantation Carcinoma
Autologous blood and red cells acute hemolysis prevention in, 478–479, AIHA in, 97, 97t, 446
for AIHA, 396–397 479t cold agglutinin syndrome in, 113t, 114
storage of, 384 AIHA after, 483–487, 484t–486t, 487f Cardiac decompensation, in severe anemia,
Autologous stem cells, for AIHA, 431–432 alloimmune thrombocytopenia after, 377
Avascular necrosis, from corticosteroids, 491–492 Cardiac surgery, cold autoantibodies in,
406–407 autoimmune thrombocytopenia after, 352–356
Azapropazone, DIIHA from, 271 491 adverse events with, 352–354, 353f
Azathioprine glycosyltransferase activity after, risk factors for, 354
for AIHA 471–472, 472f, 473t tolerance of, 354–355, 355t
in pregnancy, 349–350, 350t graft-versus-host disease after, 491. See treatment of, 355
in SLE, 444 also Graft-versus-host disease. Cardiolipin, antibodies to. See
for solid organ transplantation, graft volume of, 468–469 Anticardiolipin antibodies.
hemolysis related to, 502 hemolysis after, from cryopreserved Cardioplegia, cold, in cardiac surgery, cold
for warm antibody AIHA, 416–417 products, 499–500 autoantibodies in, 352–356, 353f,
mixed chimerism after, 487–488 355t
newly engrafted, hemolysis due to, Caspase defects, in autoimmune
474–475, 474f, 475f lymphoproliferative syndrome,
B antigen. See also ABO blood group. passenger lymphocyte syndrome after, 112
adsorption of, in bystander immune 462–463, 464t, 465, 466f, 467 Castleman’s disease, AIHA in, 113
hemolysis, 362–363, 363f pure red cell aplasia after, 476–477, CAT (column agglutination tests), for DAT-
autoantibodies to, 238, 251–253 476f–478f negative AIHA, 332
transfer of, in group O blood transfusion, Boorman, Kathleen, 21 Cataract, from corticosteroids, 408
566 Brill, I.C., 15 CD11b/CD19 (complement receptor 3), on
B lymphocytes Brule, M., 13–14 macrophages, 144f, 145
depletion of, rituximab for, 419–423, 420f, Bystander immune hemolysis, 159–160, CD16 (FcγRIII), on macrophages, 143–144,
422f 358–364 143f
intravenous Ig effects on, 425 alloimmunization in, 364 CD20, rituximab targeting, 419–423, 420f,
after transfusions, 338–339 antibody-antigen complex adsorption in, 422f
Babesiosis, AIHA in, 109–110 363 CD32 (FcγRII), on macrophages, 142–143,
Bacille-Calmette Guèrin vaccination, warm clinical settings for, 359–361, 359t 143f
antibody AIHA after, 98 complement in, 358–362, 361t CD35 (complement receptor 1), on
Band 6 protein, autoantibodies to, 244 concept of, 358 macrophages, 144–145, 144f
Banti, Guido, 15, 16f cross-reacting alloantibodies in, 364 CD52, monoclonal antibodies to, for AIHA,
Bartonella henselae infections, AIHA in, 110 definition of, 358 in chronic lymphocytic leukemia,
Basophilic stippling, in peripheral blood drug-induced, 364 444
film, 53 examples of, 358 CD64 (FcγRI), on macrophages, 141–142, 143f
Benzylpenicillin, DIIHA from, 263–264 historical background of, 358–359 Cefamandole
Beta-lactamase inhibitors, DIIHA from, 282 immune reactions in, 362–363, 363f chemical structure of, 302f
Betamethasone, for AIHA, in pediatric mechanisms of, 361–364, 361t, 363f DIIHA from, 295
patients, 344 passenger lymphocyte syndrome and, Cefazolin, chemical structure of, 302f
Bg antigen, autoantibodies to, 237 359, 463–464 Cefepime, chemical structure of, 302f
Bilirubin, 42–43 Cefoperazone, chemical structure of, 302f
determination of, in HDFN, 523–524, 523f Cefotaxime
excess of, 42–43 chemical structure of, 302f
in cold agglutinin syndrome, 73 C/c antigen DIIHA from, 295
in HDFN, 531, 532, 536 antibodies to, 231–233 prophylactic, after splenectomy, 414t
formation of, 140 in HDFN, 518 Cefotetan
in hemolytic anemias, 43 after hematopoietic cell chemical structure of, 302f
metabolism of, 43 transplantation, 490 DIIHA from, 295–299, 296t, 301, 302f, 303
unconjugated, 42–43 in hemolytic transfusion reactions, 543, Cefoxitin
in warm antibody AIHA, 64 544, 544t chemical structure of, 302f
Biopsy, chorionic villus, 524–526 passive transfusion of, 567 DIIHA from, 295–296
Blackwater, in hemoglobinuria, 49 after transfusions, 542 Ceftazidime, chemical structure of, 302f
Blood, collection of, for serologic testing, in incompatible transfusions, 559–560 Ceftriaxone
214 mimicking antibodies and, 242 chemical structure of, 302f
Blood count, 35–36 Calcium supplements, with corticosteroid DIIHA from, 295–297, 300–301
Blood culture, after splenectomy, 413 therapy, 406 cross-reactivity in, 301, 302f, 303
576 Index

Ceftriaxone (continued) Classification (continued) Cold agglutinin syndrome (continued)


prophylactic, after splenectomy, 414t of hemolytic anemias, 51, 52t, 53t Donath-Landsteiner antibody in,
Cell-mediated cytotoxicity, in red blood cell of paroxysmal cold hemoglobinuria, 62t, 193–195
destruction, 158 74 fatal, 440–441
Cellular assays, in DAT-negative AIHA, of secondary autoimmune hemolytic after hematopoietic cell transplantation,
333–334 anemias, 83, 83t 485–487, 486t, 487f
Cellular immune system, methyldopa Clinical manifestations, 35 immunochemistry of, 188–191
effects on, 273–275 of AIHA, in pediatric patients, 341–342, immunoglobulin G in, 321
Central nervous system, corticosteroid 341t, 342f, 342t incidence of, 71
therapy manifestations in, 408 of cold agglutinin syndrome, 71–73, 72t, in infections, 113
Cephalosporins 167–168 laboratory tests for, 72–73, 72t, 169, 170t,
DIIHA from, 293–303 of delayed hemolytic transfusion 182–188, 183t, 184t, 186t–187t,
case reports of, 298–301 reactions, 556, 557f, 557t 220, 222–223, 222t, 226, 227t
chemical structures and, 264, 264f, 302f of DIIHA, 290, 296–297, 296t, 297t in lymphoma, 93, 93f, 446
clinical features of, 296–297, 296t, 297t of graft-versus-host disease, 481 in malaria, 109
cross-reactivity in, 301, 302f, 303 of HDFN, 530–531, 531t molecular analysis of, 188–191
DATs in, 293–294 of hemoglobinuria, 49 in Mycoplasma pneumoniae infections,
epidemiology of, 295–296, 295t, 296t of hemolytic transfusion reactions, 556, 100–101, 169, 446
immune response in, 264, 264f 557f, 557t versus paroxysmal cold hemoglobinuria,
protein absorption in, 280–281, 281f of intravascular hemolysis, 48 194–195, 194t
serologic tests in, 297–301, 309 of mixed cold and warm antibody AIHA, in pediatric patients, 342, 342t
red blood cells treated with, for 63f, 79, 80t, 81 peripheral blood film in, 53–55, 57f
laboratory testing, 304 of paroxysmal cold hemoglobinuria, prognosis for, 73
Cephalothin 73–78 in rubella, 106, 107t
chemical structure of, 302f of passenger lymphocyte syndrome, 460, seasonal hemolysis in, 433, 435, 435f
DIIHA from, 279–281, 281f, 293–294 460f, 460t, 503 secondary, 113–114, 113t, 114t
CFAC test. See Complement fixation of secondary AIHA, 82–113 sex distribution of, 71
antibody consumption test. of warm antibody AIHA, 63–66, 64t symptoms and signs of, 71–72, 71t
Chauffard, Anatole, 8–9, 9f, 13–14 CLL. See Leukemia, chronic lymphocytic. transfusions for
Chemiluminescence test, in HDFN, 528 Clostridium perfringens septicemia, after compatibility testing for, 389–391, 390t
Chemotherapy, DIIHA from, 271, 281, hematopoietic cell transplantation, warm blood in, 395
291–292 500 washed RBCs for, 396
Chickenpox, AIHA in, 104, 106 Clough, M.C., 25 treatment of, 73, 433–441
Chido-Rogers blood group, Ge antigen of, Coagulation factor transfusions, cold avoidance in, 433, 435, 435f
autoantibodies to, 236, 245, 250, alloantibody transfer in, 567 corticosteroids in, 435–436
251, 251t Cold danazol in, 440
Chimerism. See also Microchimerism. avoidance of failure of, 440–441
definition of, 487 in cold agglutinin syndrome, 433, 435, guidelines for, 447–448
mixed, after hematopoietic cell 435f, 447 immunosuppressive therapy in,
transplantation, 487–488, 488f in Mycoplasma pneumoniae infections, 436–439, 437t
Chlorambucil 446 intravenous Ig in, 440
for AIHA in paroxysmal cold hemoglobinuria, in lymphoma, 446
in chronic lymphocytic leukemia, 443, 441 in Mycoplasma pneumoniae infections,
444 hemoglobinuria associated with. See 446
in pregnancy, 350t Paroxysmal cold options for, 402t
for cold agglutinin syndrome, 436–437, hemoglobinuria. plasma exchange in, 439
437t, 447–448 Cold agglutinin syndrome, 61, 62t, 71–73, rituximab in, 420
for warm antibody AIHA, 416 72t splenectomy in, 439–440, 440t
2-Chlorodeoxyadenosine, for AIHA, in age distribution of, 71 transfusions in, 389–391, 390t, 395, 396
chronic lymphocytic leukemia, 444 antibody characterization in, 182–188, in Waldenström’s macroglobulinemia,
Chloroquine, for IgG autoantibody 183t, 184t, 186t–187t 113–114, 113t
dissociation, 216–217 antibody specificity in, 245–253, 246t, versus warm antibody AIHA, 325
Chlorpropamide, DIIHA from, 270, 271f 251t, 252t warm antibody AIHA with. See Mixed
Chorionic villus biopsy, 524–526 autoagglutination in, 168, 168f autoimmune hemolytic anemia.
Chromic chloride method, for coupling benign antibodies in, versus in vivo Colectomy, in ulcerative colitis, AIHA
proteins to RBCs, 210–211 hemolysis, 183–188, 183t, 184t, remission after, 84, 88
Chromium, radioactive, studies with, 24 186t–187t Colitis, ulcerative, secondary AIHA in, 84,
Chronic granulomatous disease, Kell in cancer, 113t, 114 85t–89t
system autoantibodies in, 237 in cardiac surgery, 352–356 Collagen vascular diseases, cold agglutinin
Chronic lymphocytic leukemia. See adverse events with, 352–354, 353f syndrome in, 113t
Leukemia, chronic lymphocytic. versus asymptomatic cold Colton blood group, antibodies to, in
Cimetidine, DIIHA from, 293 autoantibodies, 354–355, HDFN, 519t
Ciprofloxacin, prophylactic, after 355t Column agglutination tests, for DAT-
splenectomy, 414t risk factors for, 354 negative AIHA, 332
Circulating reticulocyte maturation time, treatment of, 355 Common variable immunodeficiency,
36–37, 37f–39f case report of, 186–188, 186t–187t AIHA in, 111, 111t
Cisplatin, DIIHA from, 281, 283 clinical manifestations of, 71–73, 71t, Compatibility testing, before transfusions
Cladribine 167–168 for cold agglutinin syndrome, 389–391,
for cold agglutinin syndrome, 441 course of, 73 390t
DIIHA from, 292 differential diagnosis of, 176, 182–191, for cold-antibody AIHA, 389–391, 390t
Classification 183t, 184t, 186t–187t for paroxysmal cold hemoglobinuria,
of autoimmune hemolytic anemias, direct antiglobulin test in, 176 391
61–62, 62t, 201 discovery of, 24–26 in vivo, 394–395
Index 577

Compatibility testing, before transfusions Complement fixation antibody Cyst(s), dermoid, ovarian, AIHA in, 83–84,
(continued) consumption test (continued) 85t
for warm antibody AIHA, 378–389, 379t, for Ig measurement (continued) Cytokines, in hemolytic transfusion
381t–385t, 386f, 387t, 388t in small amounts, 321–329, 322t, 323t, reactions, 556–557
alloantibody detection in, 379–385, 325t, 328f Cytomegalovirus infections, AIHA in,
381t–384t Complement receptor 1, soluble, for warm 102–103
alloantibody incidence in, 379, 379t antibody AIHA, 424 Cytopenia
autoantibody specificity in, 385–387, Congenital hemolytic anemias, 343 in AIHA, in parvovirus B19 infections,
385t, 386f versus acquired hemolytic anemia, 5, 7, 106–107
blood selection guidelines in, 387, 13–14, 15f, 16f in autoimmune lymphoproliferative
387t definition of, 34 syndrome, 112
frequency of, 387–388 Coombs, Robin R.A., 20–21, 23, 23f DIIHA and, 275–276, 275f, 276f, 297–301
least incompatible units for, 386–387 Coombs test. See Antiglobulin (Coombs) after hematopoietic cell transplantation,
phenotype matching in, 388–389, 388t test; Direct antiglobulin test; 483–484, 490–491
red blood cell phenotyping and Indirect antiglobulin test. in warm antibody AIHA, 68
genotyping in, 378–379 Cordocentesis, in HDFN, 524–526 Cytotoxicity, of macrophages, in red blood
Compensated hemolytic disease, definition Coronary arteries, agglutination in, in cell destruction, 146–148, 148t
of, 33 cardiac surgery, 352–353
Complement Corrected reticulocyte count, 36
actions of, 134 Corticosteroids
activation of, in transfusion reactions, for AIHA, 322, 323t D antigen
556, 558 in chronic lymphocytic leukemia, 444 antibodies to, 231–234
in AIHA, in systemic lupus DAT-negative, 334 in HDFN, 517–520
erythematosus, 94, 95t intravenous Ig with, 428 in hemolytic transfusion reactions, 543,
bound to red blood cells in pediatric patients, 343 544t
antiglobulin serum containing, in pregnancy, 346, 349, 350t, 351t likelihood of, 526t
standardization of, 211–212 in SLE, 444 passive transfusion of, 567
autoantibodies and, 233, 234 for cold agglutinin syndrome, 435–436, transfusion of, hemolysis after, 496
determination of, 218 446–448 in autoadsorption technique, 383, 383t
in healthy individuals, 205–207 for mixed AIHA, 81 in hematopoietic cell transplantation,
in hospitalized patients, 208 for secondary AIHA, in ulcerative colitis, passenger lymphocyte
characteristics of, 134, 136t, 139t 84 syndrome and, 461–462
defects of, AIHA in, 111t for sickle cell hemolytic transfusion mimicking antibodies and, 242
detection of, in direct antiglobulin test, reaction syndrome, 555 Dacie, John, 19, 21, 21f, 22f
170–172, 171t for warm antibody AIHA, 401–407, 447 Dameshek, William, 16, 17f
inhibitors of, for warm antibody AIHA, adverse effects of, 405–407, 406t Danazol
424, 425f alternate-day, 404 for cold agglutinin syndrome, 440
lowering of, in washed RBC transfusion, high-dose, 402–403 for warm antibody AIHA, 423–424, 447
396 initiation of, 401–402, 403t DAT. See Direct antiglobulin test.
in malaria, 108 long-term results of, 403–404 Dedichen, H.G., 19
receptors for, on macrophages, 144–145, mechanisms of action of, 404–405, 405t Deep venous thrombosis
144f plasma exchange with, 430–431 from intravenous Ig, 429
in red blood cell destruction, 134–141, subsequent management with, 403 in warm antibody AIHA, 65
155–156 Creatine, red cell, 50 Delayed hemolysis
activation unit in, 134, 136, 136f, 137f Creite, Adolph, 4, 4f bystander immune hemolysis and,
alternative activation pathway of, Crosby, William H., 7, 8f 359–360
138–140, 138t–140t CRs (complement receptors), on after hematopoietic cell transplantation,
classical activation pathway of, macrophages, 144–145, 144f 480, 480f, 493–494
134–138, 135f–137f Cryoglobulinemia, in cold agglutinin after solid organ transplantation, 503
components of, 134, 135f, 136–138, 136f, syndrome, 72 Delayed hemolytic transfusion reactions
137f Cryoprecipitate, alloantibody transfer in, 567 versus AIHA, 356–357
with IgG, 152–154 Culture, blood, after splenectomy, 413 AIHA after, 356–357
with IgG sensitization, 152–154 Cyclophosphamide autoantibody specificity and, 357
with IgM sensitization, 154–155 for AIHA bystander immune hemolysis and,
innocent bystander, 159–160, 358–364, in chronic lymphocytic leukemia, 444 359–360
361t in pediatric patients, 343–344 clinical features of, 556, 556t, 557f
mannose-binding lectin activation in pregnancy, 350t versus delayed serologic transfusion
pathway of, 135f, 140 in SLE, 444 reactions, 545–547, 546t
membrane attack complex in, 137–138, for cold agglutinin syndrome, 436–437 before hematopoietic cell transplantation,
137f for stem cell ablation, without stem cell 473–474
by reactive hemolysis, 159–160 rescue, 432–433 in sickle cell disease, 547, 549–551
receptors in, 144–145, 144f for warm antibody AIHA, 416–418 with undetectable antibodies, 564
recognition unit in, 134, 135f–137f, 136 plasma exchange with, 430–431 Delayed hemolytic transplantation
terminology of, 134 Cyclosporine reactions
in vivo effects of, 140–141, 141t, 142f for AIHA hematopoietic cell transplantation,
without Ig sensitization, 154–156 in chronic lymphocytic leukemia, 444 473–474
sensitization to, DAT results in, 206–207 in pregnancy, 350, 350t solid organ, 503
in WAIHA serology, 178, 178f for solid organ transplantation, Delayed serologic transfusion reactions,
Complement fixation antibody hemolysis related to, 502 versus delayed hemolytic
consumption test for transplantation preconditioning, transfusion reactions, 545–547,
for AIHA, in pregnancy, 348, 348f, 348t passenger lymphocyte 546t
for Ig measurement syndrome and, 467–468 Delivery, preterm, for HDFN, 533
in hemoglobinopathies, 325–327, 325t for warm antibody AIHA, 418 Denis, Jean-Baptiste, 2, 26–29, 28f
578 Index

Dermoid cysts, ovarian, AIHA in, 83–84, 85t Differential diagnosis (continued) Direct antiglobulin test (continued)
Dexamethasone of immune hemolytic anemias versus indirect antiglobulin test, in
for AIHA, in chronic lymphocytic (continued) alloantibody detection, 379
leukemia, 444 cold response, 167–168 in low-affinity autoantibody detection,
for warm antibody AIHA, high-dose, 403 drug-induced, 168 213
Di antigen, autoantibodies to, 235 hemoglobinemia and hemoglobinuria methyldopa effects on, 150, 156–157, 157f
Diagnosis, 33–60. See also Differential in, 168–169 in mixed AIHA, 192t
diagnosis. laboratory tests in. See Laboratory negative
confirmation of, 58 test(s). in AIHA, 208–214. See also Acquired
definitions involved in, 33–35, 34f, 35f in Mycoplasma pneumoniae infections, autoimmune hemolytic
laboratory tests for. See Laboratory 169 anemia(s), negative DAT in.
test(s); specific tests. red blood cell morphology in, 169 in pregnancy, 347–349, 348f, 348t
tentative, for cause, 51–58, 52t, 53t, of mixed autoimmune hemolytic anemia, in red blood cell-bound complement,
54f–57f, 58t 191 205–206
Dialysis, after hematopoietic cell of paroxysmal cold hemoglobinuria, 168, in red blood cell-bound IgG, 204–205
transplantation, 500 176, 191, 193–196, 194t in paroxysmal cold hemoglobinuria, 176
Diamond, Louis K., 22f of reticulocytosis, 42 in passenger lymphocyte syndrome, 461,
Diclofenac, DIIHA from, 272 of systemic lupus erythematosus, 177 461t, 470, 502
Diego blood group of transfusion reactions, 168 positive
antibodies to, in HDFN, 519t DiGeorge anomaly, AIHA in, 111 blood group determination in, 214–215
Di antigen of, autoantibodies to, 235 Diglycoaldehyde (INOX), DIIHA from, 281 causes of, 203–204
Wr antigen of, autoantibodies to, 234–236 DIIHA. See Drug-induced immune in hospitalized patients, 207
Differential diagnosis, 51, 53t hemolytic anemia. phenotyping in, 215–217
of ABO incompatibility, in hematopoietic Diisopropylfluorophosphonate, in seemingly healthy individuals,
cell transplantation, 493–494 phosphorus-labeled, 24 206–207, 207t, 321
of alloantibody-induced hemolytic Dilution technique, for alloantibody significance of, 169–170, 171t, 203,
anemia, 168 detection, 380, 381t 207–208
of autoimmune hemolytic anemias, Dimethylsulfoxide, in hematopoietic cell potentiators in, 215
170–172, 171t, 191, 193–195, 194t, transplant products, 499–500 predictive value model for, 170, 171t
217–224 Diphtheria-pertussis-tetanus vaccination, principles of, 202–203, 202f, 202t
antibody characterization in, 177–178 warm antibody AIHA after, 98 reagents for, 203
antibody presence in, 218–224, 221t, Direct antiglobulin test, 58, 58t results of, versus antigen site saturation
222t in AIHA, 170–174, 171t, 172t, 173f, 174f, with IgG, 151, 151t
antibody specificity in, 224 175t, 176t sensitivity of, 321
cell sensitization considerations in, in cytomegalovirus infections, Super Coombs tests and, 332–333, 332t,
217–218 102–103 333t
cold agglutinin titer for, 220, 222–223, in HIV infection, 103–104 uses of, 202, 202t
222t in Hodgkin’s disease, 90, 92 in warm antibody AIHA, 68, 174, 176,
Donath Landsteiner test for, 223–224 in infectious mononucleosis, 102 176t, 404–405, 405t
after hematopoietic cell in leukemia, 90 Direct polybrene test, 209–210, 332, 332t,
transplantation, 492–500, 493t in malaria, 108–109 563–564
protein presence in, 218 mixed, 79, 80t, 81 Disseminated intravascular coagulation,
serum screen for, 219–220, 221t in ovarian tumors, 84 after transfusions, 392, 557, 558
warm, 174, 176, 178–182, 179t, 181t in parasitic diseases, 109–110 Dithiothreitol, in compatibility testing, 390
of cold agglutinin syndrome, 176, in rubella, 106 Diuresis, for hemolytic transfusion
182–191, 183t, 184t, 186t–187t in systemic lupus erythematosus, 95t reactions, 558
of drug-induced immune hemolytic in ulcerative colitis, 86t–88t Dizziness, in warm antibody AIHA, 63
anemia, 168, 176–177, 496 after vaccination, 99 dl (deleted) determinants, autoantibodies
of hemoglobinemia, 168–169 in varicella, 104, 106 to, 232, 232t, 233, 235, 235t
of hemoglobinuria, 168–169 in autoimmune lymphoproliferative DNA typing, of red blood cells, 379
of hemolysis, after hematopoietic cell syndrome, 112 Dodd, Barbara, 21, 23
transplantation, 492–500, 493t in cold agglutinin syndrome, 176 Donath, Julius, 12–13, 14f
AIHA, 493t, 494–496, 497t controls in, 215 Donath-Landsteiner antibody
in Clostridium perfringens septicemia, in delayed hemolytic transfusion in cold agglutinin syndrome, 193–195
500 reactions, 357 in paroxysmal cold hemoglobinuria, 77,
cryopreserved donor products and, in differential diagnosis, 169–170, 171t, 78, 191, 193–195, 194t
499–500 217–218 Donath Landsteiner test, 223–224
DIIHA, 496 in DIIHA, 171, 171t, 176–177, 278–283, Doppler studies, in HDFN, 524
dimethylsulfoxide and, 499–500 278f, 279t, 281f, 282t, 283t, Doughnut hypothesis, of red blood cell
in hemodialysis, 500 288–289 destruction, 137
major ABO blood group errors in Drug(s)
incompatibility, 493–494, 493t false-negative results from, 204 hemolytic uremic syndrome from, 499
in microangiopathic hemolytic anemia, false-positive results from, 203 thrombotic thrombocytopenia purpura
496–499, 498t false-negative, 204, 332 from, 499
minor ABO blood group false-positive, 203–204 Drug-induced immune hemolytic anemia,
incompatibility, 493 in HDFN, 517, 531, 535–536 62, 62t, 261–317
of hemolytic anemias, 51, 53t hemolysis rate versus antibody agents causing, 261–263, 262t
of immune hemolytic anemias. See also concentration and, 320–321, from alpha-interferon, 293
Differential diagnosis, of 320f, 320t, 321f from beta-lactamase inhibitors, 282
autoimmune hemolytic in hemolytic transfusion reactions, bystander immune hemolysis in, 361, 364
anemias. 567–568 from carbimazole, 268
allobody-induced, 168 immunoglobulin A detection in, 172 from cephalosporins, 293–303, 294t–297t,
autoagglutination in, 168, 168f immunoglobulin M detection in, 172 302f
Index 579

Drug-induced immune hemolytic anemia E/e antigen (continued) Evans’ syndrome (continued)
(continued) in autoadsorption technique, 383, 383t treatment of, 344
chemical structures causing, 263, 263f autoantibodies to, 231–233, 385–386, 386f in chronic lymphocytic leukemia, 444
from chemotherapy, 271, 281, 291–292 case report of, 324–325 corticosteroids in, 402
from chlorpropamide, 270, 271f in delayed hemolytic transfusion cyclosporine in, 419
clinical features of, 290 reactions, 357 danazol in, 424
diagnosis of, 290, 303–310 in older red blood cells, 243 plasma exchange in, 430
from diclofenac, 272 in younger red blood cells, 244 rituximab in, 420–422
differential diagnosis of, 168, 176–177 in incompatible transfusions, 559–560 stem cell transplantation in, 431–432
from diglycoaldehyde, 281 mimicking antibodies and, 241, 242 vinca alkaloids in, 417
direct antiglobulin test in, 171, 171t, Early erythroid progenitors, autoantibodies after umbilical cord stem cell
176–177 to, 244 transplantation, 487
drug-dependent antibodies in, 284–287, Eason, J., 12–13 in warm antibody AIHA, 67–68
284f, 285f Echinococcosis, AIHA in, 110 Extravascular hemolysis
drug-independent antibodies in, 287–288, Edema, pulmonary, from intravenous Ig, definition of, 34–35, 34f, 35f, 133
287t, 288t 429 versus intravascular hemolysis, 34–35
after hematopoietic cell transplantation, EGA kit, for IgG autoantibody dissociation, red blood cell destruction in, 151–158
496 215–216 Eye, corticosteroid therapy manifestations
historical background of, 261 Ehrlich, Paul, 10–11, 12f in, 408
in HIV infection, 104 ELAT (enzyme-linked antiglobulin test),
immune response in, 263–278 179–180, 327–328
autoantibodies in, 272 Elliptocytosis, 53, 56f
in cytopenia, 275–276, 276f Embolism, pulmonary Factor(s), in complement activation, 138,
drug characteristics and, 263, 263f, 264f, in AIHA, in HIV infection, 104 139t
265t from transfusions, 393 Factor VIII concentrate, alloantibody
hapten hypothesis of, 264f, 265–266, in warm antibody AIHA, 65 transfer in, 567
278–279, 278f En antigen, autoantibodies to, 234–236, 244 Factor IX, alloantibody transfer in, 567
immune complex hypothesis of, Encephalomyelitis, disseminated, AIHA in, Familial autoimmune hemolytic anemia,
267–272, 267f, 271f, 272t, 279, 113 351–352, 352t
279t Endotoxin, in complement activation, 138 Fanconi’s anemia, bone marrow
methyldopa-induced, 272–275, 273f Enzyme-linked antiglobulin test, 179–180, transplantation in, AIHA after, 484
new concepts of, 275–276, 276f 327–328 Fas ligand defects, in autoimmune
penicillin, 266–267, 278–279, 278f Epstein-Barr virus infections, AIHA in, lymphoproliferative syndrome,
unifying hypothesis of, 276, 277f, 278 101–102, 101t, 446–447 112
incidence of, 261 Ernstene, A.C., 25 FcγR, on macrophages, 141, 143f
laboratory tests for, 288–290, 290f, Erythroblasts, 54 FcγRI (CD64), on macrophages, 141–142,
303–310 in warm antibody AIHA, 66 143f
from levodopa, 292 Erythrocyte(s). See Red blood cell(s). FcγRII (CD32), on macrophages, 142–143,
mechanisms of, 278–283 Erythrocyte adenylate kinase test, 50 143f
immune complex, 279, 279t Erythrocyte membrane oral treatment, for FcγRIII (CD16), on macrophages, 143–144,
penicillin-type, 266–267, 278–279, 278f HDFN, 534–535 143f
protein absorption, 279–283, 281f, 282t, Erythroid hyperplasia, 50 Fertility impairment, from
283t Erythromycin, red blood cells treated with, immunosuppressive therapy, 418
from methyldopa. See under Methyldopa. for laboratory testing, 305 Fetoscopy, in HDFN, 524–526
from nomifensine, 270, 293 Erythrophagocytosis Fetus
from nonsteroidal anti-inflammatory in AIHA, 327 blood group of, determination of, 526,
drugs, 292–293 in DAT-negative AIHA, 333 526t
from penicillin. See under Penicillin(s). in differential diagnosis, 169 hemolytic disease of. See Hemolytic
from procainamide, 274–275, 292 in paroxysmal cold hemoglobinuria, 76, disease of the fetus and
from purine analogs, 291–292 77f, 195 newborn.
from quinidine, 268–272, 271f, 272t in warm antibody AIHA, 66 immunosuppressive therapy effects on,
from rifampicin, 270 Erythropoiesis 418
serologic findings in, 283–290, 284f–286f, delayed, after hematopoietic cell transfusions for, in HDFN, 524, 532–533
287t, 288t, 290f, 303–310 transplantation, 479–480 Fever
from streptomycin, 270 rate of (reticulocyte production index), in AIHA
treatment of, transfusions in, 291 37–38, 41–42, 41t in HIV infection, 104
Druitt, R., 25 suppression of, in sickle cell disease, in infectious mononucleosis, 101
Duffy blood group 553–555, 554f in warm antibody AIHA, 63
antibodies to, in HDFN, 519t in warm antibody AIHA, 69 Fifth disease. See Parvovirus B19 infections.
autoantibodies to, 237–238 Erythropoietin, for delayed erythropoiesis, Fisher, Ronald A., 22f
in incompatible transfusions, 560 after hematopoietic cell FK506 (tacrolimus)
Dyspnea, in warm antibody AIHA, 63 transplantation, 479, 480, 480f for graft-versus-host disease prevention,
Escherichia coli infections, after splenectomy, passenger lymphocyte
412 syndrome and, 468
Ethylenediaminetetraacetic acid for solid organ transplantation,
E/e antigen for blood sample collection, 214 hemolysis related to, 502
antibodies to for IgG autoantibody dissociation, Fl antigen, autoantibodies to, 250, 251, 251t
after hematopoietic cell 215–216 Flow cytometry
transplantation, 490 Evans’ syndrome for IgG determination on red blood cells,
in hemolytic transfusion reactions, 543, laboratory tests in, 342–343 150, 151t, 174, 176t, 208–209
544, 544t in pregnancy, 347 for red blood cell survival, 50
after solid organ transplantation, 503 recurrent, in primary immunodeficiency for sickle cell disease, 326–327
after transfusions, 542 diseases, 111 for warm antibody AIHA, 180
580 Index

Fludarabine Graft-versus-host disease (continued) Hematopoietic cell transplantation


for AIHA, in chronic lymphocytic after hematopoietic cell transplantation, (continued)
leukemia, 443, 444 481–483, 482f, 486 AIHA after (continued)
for cold agglutinin syndrome, 437 incidence of, 482, 482f microangiopathic, 496–499, 498t
DIIHA from, 291–292 pathophysiology of, 481 in severe combined immunodeficiency,
Fresh-frozen plasma, transfusion of, prevention of, passenger lymphocyte 111–112
autoantibody development from, syndrome and, 467–468 alloantibody formation in, 459–460
337 after solid organ transplantation, 505, non-ABO, 488–490, 488t
Functional cellular assays 505f alloimmune thrombocytopenia after,
acquired AIHA and, negative DAT in, Granulocyte transfusions, alloantibody 491–492
333–334 transfer in, 567 autoantibody formation in, 459–460
in HDFN, 526–529 Granulocytopenia, from autoimmune disease after, 490, 491
Fungal infections, from immunosuppressive therapy, 418 blood group glycosyltransferase activity
immunosuppressive therapy, 418 GVHD. See Graft-versus-host disease. persistence after, 471–472, 472f,
Fy antigens 473t
antibodies to, 238 blood group incompatibility in, 459
in hemolytic transfusion reactions, 543, graft-versus-host disease after, 481–483,
544, 544t Haemophilus influenzae infections 482f, 491
after transfusions, 542 AIHA in, 110 graft volume of, 468–469
in autoadsorption technique, 383, 383t after splenectomy, 412 immune neutropenia after, 492
in incompatible transfusions, 559–560 vaccines for, 413 immune thrombocytopenia after, 490–491
Hairy cell leukemia, AIHA in, 446 leukopenia after, 490–491
Haptens, DIIHA from, 263–266, 278–279, major ABO incompatibility in, 473–478
278f delayed hemolytic reactions in,
G antigen Haptoglobin, 43–45 473–474
autoantibodies to, 233 after blood transfusion, 44–45 differential diagnosis of, 493–494
mimicking antibodies and, 242 clinical experiences with, 44, 45f, 45t isohemagglutinin persistence after,
Galen, on hemolytic anemia, 2 deficiency of, 44 474–476, 474f, 475f
Galfenine, DIIHA from, 271 hemoglobin complex with, 44, 140 management of, 478–481, 479t, 480f,
Gangrene in hemolysis, 44, 45f, 45t 481f
in cold agglutinin syndrome, 72 normal levels of, 43, 44 pure red cell aplasia in, 476–477,
in Mycoplasma pneumoniae infections, 100 prosthetic heart valves and, 44 476f–478f
after splenectomy, 412 structure of, 43 minor ABO incompatibility in, 460–468,
Gardner, W.J., 25 Harvey, William, 2, 26 460f, 460t
Gastrointestinal system, complications of, in Hayem, Georges, 5, 7, 7f bystander hemolysis in, 463–464
corticosteroid therapy, 408 HD antigens, as early name for Pr antigens, differential diagnosis of, 493
Gd antigen, autoantibodies to, 250, 251, 251t 249 graft-versus-host disease prophylaxis
Ge antigens, antibodies to, 236, 245, 543 HDFN. See Hemolytic disease of the fetus in, 467–468
Gel centrifugation test, in warm antibody and newborn. isohemagglutinin persistence after, 473
AIHA, 180 Heart, transplantation of. See management of, 468–471, 470t
Genotyping, of red blood cells, for Transplantation, solid organ. massive hemolysis in, 463–467,
transfusions, 378–379 Heart failure 464f–466f
Gentamycin, prophylactic, after in volume overload, in transfusions, 392 passenger lymphocyte syndrome in,
splenectomy, 414t in warm antibody AIHA, 63 467, 469–470, 470t, 493
Gerbich blood group, antibodies to, in Heart valves, prosthetic serologic findings in, 461–463, 461t
HDFN, 519t haptoglobin levels with, 44 transfusion requirements in, 463, 464t
Gilbert’s syndrome lactic dehydrogenase levels with, 45, 46f, mixed chimerism after, 487, 488, 488f
hyperbilirubinemia in, 43 46t, 47f relapse after, ABO blood group and,
Ig in, 322, 322t Heinz bodies, after splenectomy, 416 481–483, 482f
Glaucoma, from corticosteroids, 408 Hematocrit types of, 459
Globoside blood group, antibodies to, in in hemolytic anemia, 41t Hematuria, versus hemoglobinuria, 49
HDFN, 519t in intravascular hemolysis, 48 Hemodialysis, after hematopoietic cell
Glucocorticoids. See Corticosteroids. normal, 41t transplantation, 500
Glucose metabolism, corticosteroid effects in red blood cell life span calculation, 38 Hemoglobin
on, 408 in reticulocyte maturation time in AIHA, in ovarian tumors, 85t
Glycine, for IgG autoantibody dissociation, calculation, 37, 38f, 39f bovine, preparations of, as red blood cell
215–216 in warm antibody AIHA, 66 substitutes, 395–396
Glycophorins, autoantibodies directed Hematopoiesis. See also Erythropoiesis. cell-free, as red blood cell substitutes,
against, 244–245 suppression of, after hematopoietic cell 396
in cold agglutinin syndrome, 249–250 transplantation, 475–476, 475f in cold agglutinin syndrome, 73
specificity of, 234–236, 235t Hematopoietic cell transplantation. See also cord, in HDFN, 532
in warm antibody AIHA, 181, 181t Stem cell transplantation; specific haptoglobin complex with, 44, 140
Glycoproteins, red cell, chemistry of, type, eg Bone marrow in mixed AIHA, 80t
248–249 transplantation. in paroxysmal cold hemoglobinuria,
Glycosyltransferase, of ABO blood group AIHA after, 483–487, 484t–486t, 487f 75–76
antigens, in hematopoietic cell in Clostridium perfringens septicemia, polymerized, as RBC substitute, 396
transplantation, 471–472, 472f, 500 in warm antibody AIHA, 66
473t cryopreserved products and, 499–500 Hemoglobinemia
Graft-versus-autoimmunity effect, in differential diagnosis of, 492–500, 493t, from anti-D, 496, 497t
allogeneic stem cell 494–496, 497t differential diagnosis of, 168–169
transplantation, 432 dimethylsulfoxide and, 499–500 in intravascular hemolysis, 48–49
Graft-versus-host disease drug-induced, 496 after transfusions, 392
clinical features of, 481 in hemodialysis, 500 in warm antibody AIHA, 64
Index 581

Hemoglobinopathies. See also Sickle cell Hemolytic disease of the fetus and newborn Historical concepts (continued)
disease. (continued) red cell agglutination discovery, 4–5, 4f,
CFAC test in, 325–327, 325t predicting presence and severity of 6f, 7f
Hemoglobinuria (continued) reticulocyte description, 9, 10f, 11f
acute, causes of, 48, 48t anti-dependent cellular cytotoxicity spherocyte description, 7–8, 8f
in AIHA, in infectious mononucleosis, assays in, 527 spleen and splenectomy roles, 15–16, 16f
101 antibody screening in, 521, 522t transfusions and transfusion reactions,
from anti-D, 496, 497t chemiluminescence test in, 528 26–28, 27f, 28f
causes of, 48t chorionic villus biopsy in, 524–526 History, medical, 51–53, 53t
chronic, causes of, 48t fetal blood group determination in, HIV infection. See Human
clinical features of, 49 526, 526t immunodeficiency virus infection.
in cold agglutinin syndrome, 71–72 fetoscopy in, 524–526 Hodgkin’s disease
consequences of, 49 functional cellular assays in, 226–229 AIHA in, 90, 92, 445
detection of, 49 Ig subclass determination in, 528–529 autoantibodies in, 238
differential diagnosis of, 168–169 medical history in, 523 Ig in, 322
early description of, 4 monocyte monolayer assay in, 527–528 Horror autotoxicus, 10–11, 12f, 336
versus hematuria, 49 quantitative antibody measurements Howell-Jolly bodies, after splenectomy, 416
in intravascular hemolysis, 48–49, 48t in, 521–523 Hr antigen, antibodies to, 240–242
nocturnal. See Paroxysmal nocturnal ultrasound in, 524 HTRs. See Transfusion reactions, hemolytic.
hemoglobinuria (PNH). umbilical blood sampling in, 524–526 Human immunodeficiency virus infection
paroxysmal cold. See Paroxysmal cold Rh immune prophylaxis for, 529–530 AIHA in
hemoglobinuria. severity of, in newborn, 531–532, 533t causes of, 103
after transfusions, 392 treatment of, 532–535 clinical manifestations of, 103–104, 105t
in warm antibody AIHA, 64 Hemolytic transfusion reactions. See diagnosis of, 104
Hemolysins Delayed hemolytic transfusion direct antiglobulin test in, 103–104
determination of, 223 reactions; Transfusion reactions, drug-related, 104
in warm antibody AIHA, 178 hemolytic. intravenous Ig for, 428
Hemolysis Hemolytic uremic syndrome, after laboratory testing in, 105t
acute, in hematopoietic cell hematopoietic cell transplantation, transfusions in, 392–394, 393f
transplantation, prevention of, 497–500, 498t treatment of, 104
478–479, 479t Hemopexin, 49 types of, 103
definition of, 33 Hemosiderinuria, 49–50 mixed AIHA in, 81
differential diagnosis of, after Heparin, for disseminated intravascular positive DAT results in, 207–208
hematopoietic cell coagulation, 558 Human leukocyte antigens, antibodies to
transplantation, 492–500, 493t Hepatitis, AIHA in, 107–108 in HTRs, 565
after hematopoietic cell transplantation Hepatitis B, vaccination for, warm antibody after transfusions, 338–339
delayed, 480, 480f, 493–494 AIHA after, 99 Huxham, John, 2
differential diagnosis of, 492–500, 493t Hepatitis C, AIHA in, 107 Hydatid disease, AIHA in, 110
immediate, 493 Hepatomegaly Hydrops fetalis, 517
laboratory tests for, 494 in AIHA, in infectious mononucleosis, clinical features of, 531
mechanisms of. See Red blood cell 101 treatment of, 533–534
destruction. in warm antibody AIHA, 64 ultrasonography in, 524
rate of, versus antibody concentration, Hereditary hemolytic anemias, 52t, 53t. See Hyper-immunoglobulin M syndrome,
320–321, 320f, 320t, 321f also specific types. AIHA in, 111t
reactive, 159–160, 361 definition of, 34 Hyperbilirubinemia, 42–43
Hemolytic anemia(s) Hh blood group, autoantibodies to, 238–239 in cold agglutinin syndrome, 73
acquired. See Acquired hemolytic Historical concepts, 1–31 in HDFN, 531, 532, 536
anemias. antiglobulin (Coombs) test, 20–21, 23f, Hyperhemolysis, after transfusions, for
autoimmune. See Autoimmune hemolytic 24t sickle cell disease, 359–360, 552
anemia(s). autoimmune hemolytic anemia concept, Hypervolemia, in transfusions, 392
causes of, 51, 52t 23–24 Hypothermia, in cardiac surgery, cold
congenital. See Congenital hemolytic autoimmune hemolytic anemia autoantibodies in, 352–354, 353f,
anemias. description, 11 355t
definition of, 33 blood investigations, 2, 3f, 4 Hypoxemia, in severe anemia, transfusions
differential diagnosis of, 51, 53t characterization of hemolytic anemias, for, 377, 377t
hereditary, 34, 52t, 53t 15–17, 17f
Hemolytic disease of the fetus and cold agglutinin syndrome discovery,
newborn, 517–540 24–26
ABO antibodies in, 535–536 controversies, 28–29 Idiopathic autoimmune hemolytic
antibodies causing, 518–521, 519t–521t, diagnostic tests, 11–12 anemia(s), 61
543t distinction between congenital and antiphospholipid antibodies in, 94, 96
assessment of, in fetus, 521–529 acquired hemolytic anemias, 5, secondary, 82–83, 82t
clinical features of, 530–531, 531t 7f Idiotypic determinants, Ig antibodies to, 245
description of, 517 Donath-Landsteiner discovery (1904), Igs. See Immunoglobulin(s).
epidemiology of, 517–518 12–13, 14f, 15f Ii blood group
historical background of, 517–518 early descriptions, 2, 6 antibodies to
in infants born to mothers with AIHA, immune hemolysis concept, 10–11, 12f in cold agglutinin syndrome, 245–249,
347 mechanism studies, 13–14, 15f, 16f 246t
predicting presence and severity of, osmotic fragility of cells, 8–9, 9f in Mycoplasma pneumoniae infections,
521–530 path of progress, 1–2 100–101
algorithm for, 525f radioactive chromium studies, 24 antigens of
amniotic fluid analysis in, 523–524, red blood cell survival measurements, characteristics of, 245–246
523f, 525f 17–20, 18f–22f chemistry of, 247–249
582 Index

Ii blood group (continued) Immunoglobulin A (continued) Immunosuppressive therapy (continued)


autoantibodies to, 238, 239t, 390–391 deficiency of, AIHA in, 111, 111t for warm antibody AIHA, 416–429, 447
in cold agglutinin syndrome, 188, 226, detection of, 172, 203 adverse effects of, 418
227t, 245–249, 246t, 390–391 receptors for, on macrophages, 144 cyclosporine, 418–419
Immediate hemolytic transfusion reactions, in red blood cell destruction, 152 danazol, 423–424, 425f
definition of, 541 standardized solutions of, 210 early reports on, 416–417
Immune complex(es), adsorption of, in Immunoglobulin G, 202 indications for, 417–418
bystander immune hemolysis, 363 bound to red blood cells intravenous Ig, 424–429, 425t, 426f,
Immune complex hypothesis, of drug- autoantibodies and, 233, 234 427f, 427t, 428t
induced immune hemolytic in cold agglutinin syndrome, 188, mycophenolate mofetil, 419
anemia, 267–272, 267f, 271f, 272t, 190–191 plasma exchange with, 430–431
279, 279t concentration of, versus hemolysis rate, regimens for, 417–418
Immune neutropenia 320–321, 320f, 320t, 321f rituximab, 419–423, 420f, 422f
after hematopoietic cell transplantation, in DAT-negative AIHA, 328–329, 328f subsequent reports on, 417
492 determination of, 172, 174, 208–209, vinca alkaloids, 417
after solid organ transplantation, 507 210, 218 Incompatibility, ABO blood group. See also
Immune response small amounts, 321–329, 322t, 323t, under ABO blood group, in
to A and B antigens, in bystander immune 325t, 328f hematopoietic cell transplantation.
hemolysis, 362–363, 363f in healthy individuals, 204–205, 321 definition of, 459
corticosteroid effects on, 408 uptake of, 327–328 Indirect antiglobulin test, 202, 202t, 220, 224,
to drugs. See Drug-induced immune in warm antibody AIHA, 178–180, 179t 226t
hemolytic anemia, immune catabolism of, in intravenous Ig action, for delayed hemolytic transfusion
response in. 425, 427f reactions, 357
to transfusions, 338–339 in complement activation, 134 versus direct antiglobulin test, in
Immune thrombocytopenia, after determination of, 172, 174, 208, 210, 218, alloantibody detection, 379
hematopoietic cell transplantation, 321–329, 322t, 323t, 325t, 328f for paroxysmal cold hemoglobinuria,
490–491 in eluates, 332–333 193, 195
Immune thrombocytopenic purpura, in sera, 333, 333t Infants. See Pediatric patients.
rituximab for, 420 dissociation of, 215–217 Infections
Immunization, AIHA after, 97–99 idiotypic determinants and, 245 AIHA in, 443
Immunodeficiency, primary, AIHA in, low-affinity, in DAT-negative AIHA, bystander immune hemolysis in,
110–111, 111t 329–330, 330f 360–361
Immunoglobulin(s) mimicking antibodies and, 240 in pediatric patients, 341
allotypes of, in solid organ nonspecific uptake of, in assays, 327–328 cold agglutinin syndrome in, 113
transplantation, 500, 502 receptors for, on macrophages, 141–144, in corticosteroid therapy, 408
bound to red blood cells, 204–205 143f in immunosuppressive therapy, 418
in complement activation, 138–139, 138t in red blood cell destruction, 148–152, paroxysmal cold hemoglobinuria in,
intravenous 149f 74–75
adverse effects of, 428–429 qualitative factors in, 151–152, 151t in rituximab therapy, 423
for AIHA quantitative factors in, 149–150, 149f, after splenectomy, 412–414, 413t, 414t
in chronic lymphocytic leukemia, 444 150t, 151t Infectious mononucleosis
in pediatric patients, 344 sensitization to, DAT results in, 206–207 AIHA in, 101–102, 101t, 446–447
in pregnancy, 350t subclasses of cold agglutinin syndrome in, 113
case reports of, 428 in DAT-negative AIHA, 328–329 Infertility, from immunosuppressive
for cold agglutinin syndrome, 440 in HDFN, 528–529 therapy, 418
for HDFN, 534 in warm antibody AIHA, 178–180, 179t Inflammatory mediators, in hemolytic
hemolysis after, 495–496, 497t Immunoglobulin M, 202 transfusion reactions, 556–557
mechanisms of action of, 425–426, 425t, bound to red blood cells Influenza, vaccines for, 414
426f, 427f in cold agglutinin syndrome, 188–191 Innocent bystander red blood cells,
results of, 426–428, 427t, 428t in DAT-negative AIHA, 331–332 destruction of. See Bystander
for warm antibody AIHA, 424–429, determination of, 210 immune hemolysis.
425t, 426f, 427f, 427t, 428t, 447 in warm antibody AIHA, 178–180, 179t Interferon-α
in red blood cell destruction, 148–155, in cold agglutinin syndrome, 188–191, DIIHA from, 293
149f, 151t 439 for lymphoma, 446
cytotoxicity in, 146–148, 148t in complement activation, 134 Interferon-α 2b, for cold agglutinin
lymphocyte interactions with, 158 detection of, 172, 203 syndrome, 438–439, 441
neutrophil interactions with, 159 in paroxysmal cold hemoglobinuria, 195 Interleukin(s), in hemolytic transfusion
qualitative factors in, 151–152, 151t in red blood cell destruction, 152, 154 reactions, 556–557
quantitative factors in, 149–150, 149f, standardized solutions of, 210 Interleukin-1 receptor antagonist, in
150t, 151t in warm antibody AIHA, 178–182, 179t, hemolytic transfusion reactions,
subclass differences in, 151–152, 151t 181t 556–557
variable macrophage activity in, Immunopancytopenia, in warm antibody Intravascular hemolysis
156–158, 157f AIHA, 68–69 cardinal features of, 34
Rh, for HDFN prophylaxis, 529–530 Immunosuppressive therapy. See also clinical features of, 48
subclasses of Corticosteroids; specific drugs. definition of, 34–35, 34f, 133
in antiglobulin serum, 213–214 for AIHA diagnosis of, 58
red blood cell destruction and, 151–152, in pediatric patients, 343–344 versus extravascular hemolysis, 34–35
151t in pregnancy, 349–351, 350t, 351t fatal, autoantibodies in, 238
Immunoglobulin A for cold agglutinin syndrome, 436–439, hemoglobinuria in, 48–49, 48t
bound to red blood cells 437t in infectious mononucleosis, 102
in cold agglutinin syndrome, 190–191 for stem cell ablation laboratory tests for, 48–49, 48t
in DAT-negative AIHA, 330–331 before autologous transplantation, 432 red cell destruction in, 133–141
determination of, 210 without stem cell rescue, 432–433 Inulin, in complement activation, 138
Index 583

Iron turnover Kidney (continued) Laboratory test(s) (continued)


in red blood cell life span calculation, 39, transplantation of. See Transplantation, for warm antibody AIHA, 169, 170t,
41f solid organ. 178–180, 178f, 179t, 225–226,
in reticulocyte maturation time Killer cells, in red blood cell destruction, 225t, 226t, 227t
calculation, 37, 37f 158–159 Lactic dehydrogenase, 45–46, 46f, 46t, 47f,
Irradiation Klebsiella pneumoniae infections, AIHA in, 47t
of blood products, for hematopoietic cell 110 Lan antigens, antibodies to, in hemolytic
transplantation, 481, 481f Kp antigen, autoantibodies to, 236–237 transfusion reactions, 543
splenic, for warm antibody AIHA, 416 Kuessner, B., 11 Landois, Leonard, 4–5
Isohemagglutinins Kupffer cells, red blood cell destruction in, Landsteiner, Karl, 5, 5f, 6f, 7f, 12–13, 14f
after hematopoietic cell transplantation, 140 Landsteiner-Weiner blood group,
473–476, 474f, 475f antibodies to, in HDFN, 519t
removal of Laparoscopic approach, to splenectomy,
for acute hemolysis prevention, 407–408
478–479 Laboratory test(s). See also specific tests and Le antigens, antibodies to, in hemolytic
for delayed erythropoiesis prevention, types of anemia. transfusion reactions, 543, 543t, 544t
479 for AIHA Least incompatible units, in transfusion,
Iwai, S., 25 DAT-negative, 332–333, 332t, 333t 386–387
in pediatric patients, 342–343, 342t Lederer, M., 15
for antibodies, failure of, 563–567 Legionella pneumophila infections, AIHA in,
antiglobulin test as. See Antiglobulin 110
Jaundice (Coombs) test; Direct Leishmaniasis, systemic, AIHA in, 109
in AIHA antiglobulin test; Indirect Leptospirosis, AIHA in, 110
in infectious mononucleosis, 101 antiglobulin test. Leukemia
in pediatric patients, 341, 341t for autoantibody specificity, 224–225, AIHA in, 81, 112, 158–159, 446
in HDFN, 531, 536 225t, 226t chronic lymphocytic
in paroxysmal cold hemoglobinuria, 75 bilirubin measurement as, 42–43 AIHA in, 89–90, 89t, 90f, 91t
in warm antibody AIHA, 64 blood collection for, 214 incidence of, 441, 442t, 443
Jk antigens blood count as, 35–36 intravenous Ig for, 428
in allogeneic adsorption test, 384–385 blood group determination, 214–215 rituximab for, 420–422
antibodies to, 237–238 for cold agglutinin syndrome, 72–73, 72t, treatment of, 443–444, 443f
in hemolytic transfusion reactions, 543, 169, 170t, 182–188, 183t, 184t, chemotherapy for, DIIHA from,
544, 544t 186t–187t, 220, 222–223, 222t, 291–292
after solid organ transplantation, 506 226, 227t cold agglutinin syndrome in, 189–190
in solid organ transplantation, 502 confirmatory, 58 hemolysis with, cyclosporine for, 419
in autoadsorption technique, 383, 383t for differential diagnosis, 217–224 natural killer cells in, 334
in hematopoietic cell transplantation, for DIIHA, 303–310 transfusions for, AIHA after, 337
passenger lymphocyte carbodiimide in, 307 chronic myelogenous, hematopoietic cell
syndrome and, 462 cefotetan antibody detection, 309 transplantation for
in incompatible transfusions, 559–560 from cephalosporins, 297–301, 309 AIHA after, 483, 487
Ju antigen, autoantibodies to, 253 drug inhibition test, 310 alloimmune thrombocytopenia after,
drug solutions for, 304 491–492
drug-treated RBCs for, 304–305 immune neutropenia after, 492
immune complex method, 307–308 cold agglutinin syndrome in, 113–114,
K antigen direct antiglobulin test as. See Direct 113t, 114t
alloantibodies to, 380, 381t antiglobulin test. hairy cell, AIHA in, 446
antibodies to, 236–237 for drug-induced immune hemolytic mixed AIHA with, 81
in HDFN, 518 anemia, 288–290, 290f, 303–310 transfusions for, antibody test failure in,
in hemolytic transfusion reactions, 543, for haptoglobin, 43–45, 45f, 45t 563
544, 544t for HDFN, 520–524, 523f Leukocyte(s). See also Leukocytosis;
passive transfusion of, 567 for hemoglobinemia, 48–49 Leukopenia.
after transfusions, 542 for hemoglobinuria, 48–49 in paroxysmal cold hemoglobinuria, 76
in autoadsorption technique, 383, 383t for hemolysis, after hematopoietic cell reduction of
Kala-azar, AIHA in, 109 transplantation, 494 in hematopoietic cell transplantation,
Kawasaki disease, AIHA in, 113 for hemolytic transfusion reactions, 481, 481f
Kell blood group 567–568 for red blood cell transfusions, 396
antibodies to, 236–237, 519t, 543t for hemopexin, 49 in warm antibody AIHA, 66
in incompatible transfusions, 560 for hemosiderinuria, 49–50 Leukocytosis
Kernicterus, in HDFN, 531 for infections, after splenectomy, 413 in paroxysmal cold hemoglobinuria, 76
Kidd blood group for intravascular hemolysis, 48–49, 48t, 58 in warm antibody AIHA, 66
antibodies to, 237–238, 519t for lactic dehydrogenase, 45–46, 46f, 46t, Leukopenia
in incompatible transfusions, 559–560 47f, 47t from immunosuppressive therapy, 418
Kidney for methemalbumin, 49 in paroxysmal cold hemoglobinuria, 76
failure of for mixed AIHA, 191, 192t in warm antibody AIHA, 68
in hemoglobinuria, 49 overview of, 169, 170t Levine, Philip, 18, 20f
in hemolytic transfusion reactions, for paroxysmal cold hemoglobinuria, Levodopa, DIIHA from, 292
557–558 191, 193–195, 194t, 227, 227t Lewis blood group
from intravenous Ig, 429 for passenger lymphocyte syndrome, 493 in AIHA, in infectious mononucleosis,
in passenger lymphocyte syndrome, peripheral blood film as, 53–58, 54f–57f 102
463 for red blood cell morphology, 42 antibodies to
hemolytic uremic syndrome and, for reticulocytes, 36–42, 37f–41f, 41t for antiglobulin serum preparation, 211
497–500, 498t transfusion requirements as, 46–48, 48t passenger lymphocyte syndrome and,
hemosiderin deposition in, 49–50 value of, 50–51 462
584 Index

Li antigen, autoantibodies to, 250–251, 251t Macroglobulinemia (continued) Methyldopa, DIIHA from
Lily optical density method, in HDFN, Waldenström’s antibodies in, 287–288, 288t
523–524, 523f cold agglutinin syndrome in, 113–114, diagnosis of, 290
Lipid metabolism, corticosteroid effects on, 113t, 114t laboratory tests in, 288–290, 290f
408 mixed AIHA with, 81 mechanisms for, 273–275, 283
Liver Macrophage(s), in red blood cell positive DAT in, 150, 156–157, 157f
enlargement of destruction, 141–158 transfusions in, 397
in AIHA, in infectious mononucleosis, armed, 160–161, 160f Methylprednisolone
101 complement receptors on, 144–145, 144f for AIHA, in pediatric patients, 343–344
in warm antibody AIHA, 64 cytotoxicity of, 146–148, 148t for delayed erythropoiesis, after
failure of, in warm antibody AIHA, 64 origin of, 140, 142f hematopoietic cell
red blood cell destruction in, 140, 148 qualitative factors in, 151–152, 151t transplantation, 478
transplantation of. See Transplantation, quantitative factors in, 149–150, 149f, for warm antibody AIHA, 402–403, 417
solid organ. 150t, 151t Microangiopathic hemolytic anemia, after
Loutit, John F., 19, 23 receptors in, 141–144, 143f hematopoietic cell transplantation,
Lower, Richard, 26, 27f, 28 sensitized with complement alone, 496–499
Lu antigens 155–156 causes of, 496–497
alloantibodies to sensitized with IgA alone, 152 diagnosis of, 497–498
after hematopoietic cell sensitized with IgG alone, 148–152, 149f, differential diagnosis of, 499
transplantation, 489–490 150t, 151t incidence of, 498
after transfusions, 542 sensitized with IgG and complement, risk factors for, 498–499
antibodies to, 239 152–154 thrombotic thrombocytopenia in, 497,
in hemolytic transfusion reactions, 543, sensitized with IgM alone, 152 498t
543t sensitized with IgM and complement, treatment of, 499
Lud antigen, autoantibodies to, 251, 251t 154–155 Microchimerism
Lung variation in activity of, 156–158, 157f after solid organ transplantation, 505,
acute transfusion-related injury of, from Major incompatibility, ABO blood group. 505f, 507
intravenous Ig, 429 See also under ABO blood group, in after transfusion, 340
transplantation of. See Transplantation, hematopoietic cell Microcytes, discovery of, 5
solid organ. transplantation. Mimicking antibodies, 239–242, 241t
Lupus anticoagulant definition of, 459 Minkowski, O., 5
in AIHA, 94 Malaria, AIHA in, 108–109 Mino, P., 25
in warm antibody AIHA, 65 Malignant autoimmunity, stem cell Minor incompatibility, ABO blood group.
Lutheran blood group transplantation for, 431–432 See also under ABO blood group, in
antibodies to, in HDFN, 519t Mannose-binding lectin pathway, of hematopoietic cell transplantation.
autoantibodies to, 239 complement activation, 135f, 140 definition of, 459
LW blood group, autoantibodies to, Marrow transit time Mixed autoimmune hemolytic anemia, 61,
231–234, 253 in hemolytic anemia, 41t 79, 80t, 81
Lymphadenopathy normal, 41t age distribution of, 63f
in AIHA, in leukemia, 89 Masius, J.R., 5, 7 case reports of, 79, 80t, 81
in autoimmune lymphoproliferative Matuhasi-Ogata phenomenon, 239–242, course of, 81
syndrome, 112 241t diagnosis of, 81
in infectious mononucleosis, 101 Maturation, of reticulocytes, 36–37, 37f–39f differential diagnosis of, 191, 192t
in warm antibody AIHA, 64 Me antigen, autoantibodies to, 253 direct antiglobulin test in, 192t
Lymphocytes. See also B lymphocytes; Mean corpuscular hemoglobin laboratory tests in, 191, 192t
Passenger lymphocyte syndrome; concentration, in intravascular in pediatric patients, 342, 342t
T lymphocytes. hemolysis, 48 secondary, 81
proliferation of, in cold agglutinin Mean total reactivity of adherence and treatment of, 81, 438
syndrome, 73 phagocytosis, in DAT-negative MNS blood group
in red blood cell destruction, 158 AIHA, 333–334 antibodies to, in HDFN, 519t
Lymphocytosis, in infectious Measles vaccination, warm antibody AIHA U antigen of, autoantibodies to, 234, 235t
mononucleosis, 101 after, 99 Mollison, Patrick, 19, 22f
Lymphoma. See also Hodgkin’s disease. Mefenamic acid, DIIHA from, 292 Monoclonal antibodies
AIHA in, 92–93, 92t, 93f, 112, 446 Mei-Sai, N., 25 to CD52, for AIHA, in chronic
autoantibodies in, 238 Membrane attack complex, in complement lymphocytic leukemia, 444
cold agglutinin syndrome in, 113–114, activation, 137–138, 137f rituximab. See Rituximab.
113t, 114t, 189–190, 438 Meningitis, from intravenous Ig, 429 Monoclonal gammopathy
hematopoietic cell transplantation for, Meningococcal vaccines, 413–414 cold sensitive antibodies in, 195–196
passenger lymphocyte 2-Mercaptoethanol, in compatibility testing, Pr autoantibodies in, 250
syndrome after, 465 390 Monocyte(s)
paroxysmal cold hemoglobinuria in, 75 6-Mercaptopurine erythrophagocytosis by, in DAT-negative
Lymphoproliferative disorders. See also for AIHA AIHA, 333–334
Leukemia; Lymphoma. in pediatric patients, 344 in red blood cell destruction, 146–148,
AIHA in, 88–94, 89t, 90f, 91t, 92t, 93f, 428 in pregnancy, 350t 153, 157, 160
autoimmune, AIHA in, 112, 112t for warm antibody AIHA, 416 in warm antibody AIHA, 66
cold agglutinins in, 189 Metastasis, secondary AIHA in, 97 Monocyte monolayer assay
Methemalbumin, 49, 139 in DIIHA, 283
Methemoglobin, in urine, 49 in HDFN, 527–528
Methotrexate Mononuclear phagocyte assay, in DAT-
M antigen, antibodies to, 236, 253, 543, 544t for AIHA, in pregnancy, 350t negative AIHA, 333–334
Mackenzie, Stephen, 11 for graft-versus-host disease prevention, Mononucleosis, infectious
Macroglobulinemia passenger lymphocyte AIHA in, 101–102, 101t, 446–447
autoantibodies in, 253 syndrome and, 467–468 cold agglutinin syndrome in, 113
Index 585

Monospot test, for infectious Neutropenia Paroxysmal cold hemoglobinuria (PCH)


mononucleosis, 101 in autoimmune lymphoproliferative (continued)
Moreschi, Carlo, 21, 23, 23f, 24t syndrome, 112 chronic, 77–78, 78f
Morgenroth, Julius, 10 immune classification of, 62t, 74
Mortality after hematopoietic cell versus cold agglutinin syndrome,
in AIHA, in pediatric patients, 344 transplantation, 492 194–195, 194t
in cold agglutinin syndrome, 73, 440–441 after solid organ transplantation, 507 definition of, 196
in HDFN, 531 in warm antibody AIHA, 67, 68 diagnosis of, historical background of,
in hematopoietic cell transplantation, Neutrophil(s) 11–12
ABO blood group and, 481–483, in paroxysmal cold hemoglobinuria, 76 differential diagnosis of, 168, 176, 191,
482f in red blood cell destruction, 153, 159 193–196, 194t
in HTRs, 559–560 Neutrophilia, in warm antibody AIHA, direct antiglobulin test in, 176
in infants of mothers with AIHA, 346 66 early description of, 2
in rituximab therapy, 423 Newborn, hemolytic disease of. See erythrophagocytosis in, 195
in splenectomy Hemolytic disease of the fetus and first description of, 11–12
infectious, 412 newborn. hematologic findings in, 75–76, 77f
surgical, 411–412 nl (normal) determinants, autoantibodies idiopathic, 74–75
in transfusion reactions, 336 to, 232, 232t, 233, 235, 235t, 385 immunoglobulin M antibodies in, 195
in warm antibody AIHA, 70–71 Nocturnal hemoglobinuria. See incidence of, 73–74
Mourant, Arthur, 21, 22 Paroxysmal nocturnal laboratory tests for, 191, 193–196, 194t,
Mouse macrophage assay, in DAT-negative hemoglobinuria (PNH). 227, 227t
AIHA, 333 Nomifensine, DIIHA from, 270, 293 in lymphoma, 446
Mucocutaneous candidiasis, AIHA in, 111, Nonsteroidal anti-inflammatory drugs, in pediatric patients, 342, 342t, 343
111t DIIHA from, 292–293 prognosis for, 78
Mucocutaneous disorders, from rituximab, racial distribution of, 74
423 secondary, 74–75
Multiorgan failure, from rituximab, 423 sex distribution of, 74
Multiple myeloma Oldenburg, Henry, 28–29 symptoms and signs of, 75, 75t
AIHA in, 113 OPSI (overwhelming postsplenectomy transfusions for, 391, 395
bone marrow transplantation in, AIHA infection), 412–414, 413t, 414t treatment of, 78, 441
after, 484 Optical density measurements, of bilirubin, guidelines for, 448
Mycophenolate mofetil in HDFN, 523–524, 523f options for, 402t
for transplantation preconditioning, Osteoporosis, from corticosteroids, 406 in varicella, 106
passenger lymphocyte Ovalocytosis (elliptocytosis), 53, 56f Paroxysmal nocturnal hemoglobinuria
syndrome and, 467 Ovarian tumors, AIHA in, 83–84, 85t (PNH)
for warm antibody AIHA, 419 Overwhelming postsplenectomy infection, bystander immune hemolysis in,
Mycoplasma pneumoniae infections 412–414, 413t, 414t 360–361
AIHA in, 100–101, 446–447 Oxaliplatin, DIIHA from, 281 hemosiderinuria in, 50
cold agglutinin syndrome in, 113, 169, Oxyhemoglobin, in urine, 49 IgG in, 321
354 Partial immunoreaction, in hapten
Myelodysplastic syndromes, AIHA in, 113 hypothesis, 265
Myeloproliferative disorders, AIHA in, Parvovirus B19 infections
88–94, 89t, 90f, 91t, 92t, 93f P antigen AIHA in, 106–108
Myocardial infarction, from intravenous Ig, antibodies to, in hemolytic transfusion paroxysmal cold hemoglobinuria in, 75
429 reactions, 543, 544t reticulocytopenia in, 70
Myocardial necrosis, in cold agglutinin autoantibodies to, in paroxysmal cold Passenger lymphocyte syndrome
syndrome, in cardiac surgery, hemoglobinuria, 254 bystander immune hemolysis and, 359,
352–353 in paroxysmal cold hemoglobinuria, 75 463–464
in parvovirus B19 infections, 70 clinical setting of, 493
Pain diagnosis of, 493
in paroxysmal cold hemoglobinuria, 75 in hematopoietic cell transplantation,
N antigen, antibodies to, 236, 253 in sickle cell disease, 547–548, 555 460–468
in AIHA, in infectious mononucleosis, in warm antibody AIHA, 63 antibody sources in, 462
102 Pallor bystander hemolysis and, 463–464
Nafcillin in AIHA, in pediatric patients, 341, 341t clinical features of, 460, 460f, 460t
DIIHA from, 286 in paroxysmal cold hemoglobinuria, 75 course of, 463, 464f
red blood cells treated with, for Palpitations, in warm antibody AIHA, 63 description of, 460
laboratory testing, 305 Pancreas, transplantation of. See donor-derived antibodies in, 462–463
Natural killer cells, in red blood cell Transplantation, solid organ. graft-versus-host disease prophylaxis
destruction, 158–159, 334, 565–566 Pancytopenia and, 467–468
Necrosis, avascular, from corticosteroids, in AIHA, in parvovirus B19 infections, immune modulation in, 467
406–407 107 laboratory findings in, 460, 460f, 460t
Neisseria meningitidis infections, after after hematopoietic cell transplantation, Lewis blood group and, 462
splenectomy, 412 483–484 massive hemolysis in, 463–467,
Neonates, hemolysis in. See Hemolytic in warm antibody AIHA, 68–69 464f–466f
disease of the fetus and newborn. Pappenheim, Artur, 10, 10f monitoring of, 469–470, 470t
Neoplasia, from immunosuppressive Parasitic diseases, AIHA in, 108–110 onset of, 460
therapy, 418 Paroxysmal cold hemoglobinuria (PCH), Rh blood system and, 461–462
Neurologic disorders 61–62, 62t, 73–78 risk factors for, 460
from corticosteroids, 408 age distribution of, 63f, 74 serologic findings in, 461–463, 461t
from intravenous Ig, 429 antibody specificity in, 254 stem cell source and, 467
in severe anemia, 377 autoantibodies in, 12–13, 14f, 195–196, transfusion requirements in, 463, 464t
in warm antibody AIHA, 63 254 laboratory tests for, 493
586 Index

Passenger lymphocyte syndrome Pharyngitis, in infectious mononucleosis, Prednisone (continued)


(continued) 101 for warm antibody AIHA (continued)
in solid organ transplantation, 500–504, Phenotyping, 215–217, 218t long-term results of, 403–404
501t of red blood cells, for transfusions, subsequent management with, 403
case reports of, 503 378–379, 388–389, 388t Pregnancy
clinical features of, 503 of reticulocytes, 378–379 AIHA in, 344–351, 345f
donor-derived antibodies in, 503 Phosphatidylserine, on red blood cells, in fetal outcome in, 346, 347
factors affecting hemolysis in, 502 warm antibody AIHA, 65 incidence of, 349
Ig allotypes in, 500, 502 Phospholipids, antibodies to. See infants without hemolysis and, 347
serologic findings in, 502 Antiphospholipid antibodies and maternal findings in, 345–346
treatment of, 503–504, 504t antiphospholipid syndrome. with negative DAT, 347–349, 348f, 348t
PCH. See Paroxysmal cold hemoglobinuria. Phototherapy, for HDFN, 533, 536 neonatal hemolysis in, 347
pd (partially deleted) determinants, Physical examination, 51–53, 53t recurrent, 345, 345f, 347–348
autoantibodies, 232, 232t, 233, 235, Piperacillin, DIIHA from, 286–287 severe, 345–346
235t Placenta, ultrasonography of, in HDFN, 524 splenectomy in, 351
Pediatric patients Plasma, transfusion of, alloantibody transient, 348
AIHA in. See also Hemolytic disease of transfer in, 567 treatment of, 349–351, 350t, 351t
the fetus and newborn. Plasma exchange fetal-maternal cell transfer during, 340.
versus adults, 340 for AIHA See also Hemolytic disease of the
chronic, 341, 341t in infectious mononucleosis, 447 fetus and newborn.
clinical findings in, 341–342, 341t, 342f, in pediatric patients, 344 immunosuppressive therapy in, 418
342t for cold agglutinin syndrome, 439, 448 sickle cell hemolytic transfusion reaction
course of, 341–342, 341t, 342f, 342t before cardiac surgery, 355–356 syndrome in, 551
idiopathic, 342 for HDFN, 525, 534 Pretransplant conditioning regimen
laboratory findings in, 342–343, 342t for paroxysmal cold hemoglobinuria, 441 nonmyeloablative, pure red cell aplasia
prognosis for, 344 for warm antibody AIHA, 429–431 after, 477, 477f, 478f
rituximab for, 421–423, 422f Plasmodium infections, AIHA in, 108–109 passenger lymphocyte syndrome and,
after transfusions, 339–340 Platelet(s) 467
transient, 341, 341t abnormal levels of. See Primary immunodeficiency, AIHA in,
treatment of, 343–344 Thrombocytopenia; 110–111, 111t
in very young patients, 343–344 Thrombocytosis. Procainamide, DIIHA from, 274–275, 292
corticosteroid complications in, 408 antibodies to Promethazine, for HDFN, 534
Evans’ syndrome in. See Evans’ after hematopoietic cell Properdin, in complement activation,
syndrome. transplantation, 491 138–140, 139t
paroxysmal cold hemoglobinuria in, 74, in warm antibody AIHA, 68 Protein
343 count of, in pediatric patients, 342 coupling to red blood cells, chromic
runt disease in, 340 in paroxysmal cold hemoglobinuria, 76 chloride method for, 210–211
sickle cell hemolytic transfusion reaction transfusion of nonspecific uptake of, in direct
syndrome in, 551–552 alloantibody transfer in, 567 antiglobulin test, 203–204
splenectomy in, 343, 412 hemolysis after, 494–495 Pseudohyperkalemia, after splenectomy,
thalassemia in, hemolytic transfusion volume reduction in, 469 415
reactions in, 552 in warm antibody AIHA, 66–68 Pulmonary edema, from intravenous Ig, 429
warm antibody AIHA in, 341–344, 341t, Pneumococcal infections. See Streptococcus Pulmonary embolism
342f, 342t pneumoniae infections. in AIHA, in HIV infection, 104
in cytomegalovirus infections, 103 Poikilocytosis from transfusions, 393
thymectomy for, 433 in cold agglutinin syndrome, 73 in warm antibody AIHA, 65
Penicillin(s), DIIHA from in sickle cell disease, 55f Pure red cell aplasia
antibodies in, 266–267, 283–287, Poliomyelitis vaccination, warm antibody En autoantibodies in, 244
284f–286f AIHA after, 98, 99 after hematopoietic cell transplantation,
chemical structures of, 264f, 266, 302f Polyagglutinable red blood cells, direct 476–477, 476f–478f, 479–480
cross-reactivity in, 301, 302f, 303 antiglobulin test in, 204 rituximab for, 421–422
immune response in, 263–264, 264f, Polybrene test, direct, 209–210, 332, 332t, in warm antibody AIHA, 70
266–267 563–564 Purpura. See also Thrombocytopenic
impurities in preparations, 266–267 Polychromasia, in warm antibody AIHA, 66 purpura.
mechanisms of, 278–279, 278f, 279t PolyHeme, as RBC substitute, 396 posttransfusion, 360
serologic diagnosis of, 303–308 Pr antigens, autoantibodies to, 235–236,
Peptic ulcer disease, from corticosteroids, 249–250, 251t, 252t
408 in cold agglutinin syndrome, 188,
Perfluorochemicals, as RBC substitutes, 396 245–246, 249–250 Quinidine, DIIHA from, 268–272, 271f, 272t
Peripheral blood film, 53–58, 54f–57f, 66 Prásek, Emil, 6f
Peripheral blood stem cell transplantation Prednisolone, for AIHA
hemolysis after, from cryopreserved in pediatric patients, 344
products, 499–500 in pregnancy, 346 Race, Robert R., 20, 21, 22f
passenger lymphocyte syndrome after, Prednisone Raynaud’s phenomenon, 25
462, 465–467, 466f for AIHA versus acrocyanosis, in cold agglutinin
Phagocytosis. See also Erythrophagocytosis. in chronic lymphocytic leukemia, 443 syndrome, 72
inhibition of, in intravenous Ig action, in pediatric patients, 344 in paroxysmal cold hemoglobinuria, 78
425–426 for cold agglutinin syndrome, 435–436, RBCs. See Red blood cell(s).
in paroxysmal cold hemoglobinuria, 76, 447 RCA proteins, for warm antibody AIHA,
76f for warm antibody AIHA, 447 424, 425f
of senescent red blood cells, antibodies alternate-day, 404 Reactive hemolysis, 159–160, 361
in, 205 high-dose, 402–403 in bystander immune hemolysis,
in warm antibody AIHA, 66 initiation of, 401 359–360
Index 587

Recognition unit, in complement activation, Red blood cell(s) (continued) Reticulocyte(s) (continued)
134, 136f survival of laboratory tests for (continued)
Red blood cell(s) measurement of, 17–20, 18f–22f interpretation of, 40–42
agglutination of. See Agglutination. after transfusion, 383–384, 384t. See also red blood cell life span as, 38–39, 40f,
antigens and antibodies of. See Red blood Transfusion reactions, 41f, 41t
cell antibodies; Red blood cell hemolytic. reticulocyte count or percentage as, 36
antigens. in in vivo compatibility testing, reticulocyte maturation index as, 39
aplasia of. See Pure red cell aplasia. 394–395 reticulocyte maturation time as, 36
autologous typing of, after hematopoietic cell reticulocyte production index as, 37–38,
for AIHA, 396–397 transplantation, 471–472, 473t 41–42, 41t
storage of, for future autoadsorption washed, 396 in mixed AIHA, 80t
test, 384 Red blood cell antibodies. See also specific in paroxysmal cold hemoglobinuria, 76
clearance of, in warm antibody AIHA, antibodies. phenotyping of, 378–379
corticosteroid effects on, 405 in complement activation, 140, 141t stress (shift), 36, 39
complement bound to. See Complement, pathogenicity of, 148, 148t in warm antibody AIHA, 69–70, 69f, 69t
bound to red blood cells. Red blood cell antigens. See also specific Reticulocyte count, 36
creatine measurement in, 50 antigens. absolute, 36, 39–40, 40f
destruction of. See Red blood cell alterations of, antibodies to, 273 accuracy of, 39
destruction. in ulcerative colitis, 88 corrected, 36
early description of, 2, 3f Red blood cell destruction in hemolytic anemia, 41t
exchange transfusions of, before agglutination in, 161 interpretation of, 40–42
hematopoietic cell complement activation in, 133–141 normal, 41t
transplantation, 469 alternative pathway of, 138–140, 139t, in red blood cell life span calculation, 38,
formation of. See Erythropoiesis. 140t 39, 40f
fragmented, in peripheral blood film, 53, classical pathway of, 134–138, Reticulocyte maturation index, 39
56f 135f–137f Reticulocyte maturation time, 36–37, 37f–39f
genotyping of, for transfusions, mannose-binding lectin pathway of, Reticulocyte production index, 37–38,
378–379 135f, 140 41–42, 41t
glycoproteins of, chemistry of, 248–249 in vivo effects of, 140–141, 141t, 142t Reticulocytopenia
immunoglobulins bound to. See specific complement receptors in, 144–145, 144f in AIHA
immunoglobulins, bound to red immunoglobulin Fc receptors in, 144 in pediatric patients, 342
blood cells. “innocent bystander.” See Bystander transfusion for, 376
increased mass of, in transfusions, 378 immune hemolysis. in cold agglutinin syndrome, 73
innocent bystander, destruction of. See low-affinity Ig and, 329–330, 330f in hemolytic anemia, 42
Bystander immune hemolysis. lymphocytes in, 158 in paroxysmal cold hemoglobinuria, 76
irregularly contracted, 53, 53t macrophages in, 145–158, 145f–147f in sickle cell hemolytic transfusion
leukocyte-reduced, 396, 481, 481f armed, 160–161, 160f reaction syndrome, 549
life span of cytotoxicity of, 146–148, 148t in warm antibody AIHA, 69–70, 69f, 69t
estimation of, 38, 39, 40f, 41f, 41t qualitative factors in, 151–152, 151t Reticulocytosis
flow cytometry for, 50 quantitative factors in, 149–150, 149f, in AIHA, in parvovirus B19 infections,
mass of, excessive, in transfusions, 378 150t, 151t 107
membranes of, for HDFN, 534–535 receptors in, 141–144, 143f in cold agglutinin syndrome, 73
morphology of, 42 sensitized with complement alone, in corticosteroid therapy, for warm
in cold agglutinin syndrome, 73 155–156 antibody AIHA, 402
in differential diagnosis, 169 sensitized with IgA alone, 152 differential diagnosis of, 42
in paroxysmal cold hemoglobinuria, sensitized with IgG alone, 148–152, direct antiglobulin test in, 204
76 149f, 150t, 151t Reticuloendothelial system, 133
osmotic fragility of, 8–9, 9f sensitized with IgG and complement, methyldopa effects on, 290, 290f
packed, transfusion of 152–154 red cell destruction in, 151–158
alloantibody passive transfer in, 566 sensitized with IgM alone, 152 Rh blood group. See also specific antigens.
hemolysis after, 496 sensitized with IgM and complement, alloantibodies to, after hematopoietic cell
panel of, for alloantibody detection, 380, 154–155 transplantation, 489–490
381t variation in activity of, 156–158, 157f antibodies to
phagocytosis of. See natural killer cells in, 158–159 in DIIHA, 289–290
Erythrophagocytosis. neutrophils in, 159 in HDFN, 519t, 530
phenotyping of, for transfusions, overview of, 141, 143f in hemolytic transfusion reactions, 543t
378–379, 388–389, 388t reactive hemolysis in, 159–160 of recipient origin, 505–506
phosphatidylserine on, in warm antibody Red cell aplasia. See Pure red cell aplasia. in red blood cell destruction, 149, 149f
AIHA, 65 Relative specificity, of autoantibodies, 385, in solid organ transplantation, 502
pitting of, after splenectomy, 416 385t after transfusion, 336–337, 339
polyagglutinable, direct antiglobulin test Reticulocyte(s) autoantibodies to, 231–234, 385, 385t
in, 204 abnormal. See Reticulocytopenia; in hematopoietic cell transplantation
protein coupling to, chromic chloride Reticulocytosis. alloantibodies and, 489–490
method for, 210–211 in AIHA, in ovarian tumors, 85t passenger lymphocyte syndrome and,
removal of, from donor marrow, 478 D autoantibodies in, 243 461–462
saline-suspended, for compatibility discovery of, 9, 10f, 11f immunoglobulins of, in HDFN, 529–530
testing, 378–380, 390t fractionation methods for, 379 mimicking antibodies and, 240–242
skeletal proteins of, autoantibodies to, laboratory tests for, 36–42 phenotype determination in
244–245 absolute reticulocyte count as, 36, 39–40 in positive DAT, 215
storage of accuracy of, 39 for transfusions, 378–379
antibody reactions in, 253–254 circulating reticulocyte maturation Rheumatoid arthritis, AIHA in, 96
autoantibody specificity and, 243 time as, 36–37, 37f–39f Richter, J.M., 25
substitutes for, 395–396 corrected reticulocyte count as, 36 Rifampicin, DIIHA from, 270
588 Index

Rituximab Septicemia, Clostridium perfringens, after Specificities (continued)


for AIHA hematopoietic cell transplantation, of autoantibodies (continued)
in chronic lymphocytic leukemia, 444 500 antiphospholipid, 245
in lymphoma, 446 Serology. See Laboratory test(s). changes in, 242–243
for cold agglutinin syndrome, 437–438 Severe combined immunodeficiency in cold agglutinin syndrome, 245–253,
for mixed AIHA, 81 bone marrow transplantation for, 246t, 251t, 252t
for warm antibody AIHA, 419–423 alloantibodies after, 489 in delayed hemolytic transfusion
adverse effects of, 423 hematopoietic cell transplantation in, reactions, 357
in children, 421–423, 422f AIHA after, 111–112 drug-induced, 289–290, 290f
efficacy of, 420–421, 420f Shift (stress) reticulocytes, 36, 39 Duffy system, 238–239, 239t, 240t
RMI (reticulocyte maturation index), 39 Shock Fl antigen, 250, 251, 251t
Rosette formation, in red blood cell in fulminant hemolytic anemia, 377 Gd antigen, 250, 251, 251t
destruction, 145, 145f, 146f, 157 septic, after splenectomy, 412 glycophorins and, 234–236, 235t
Roth, G., 25 Sia antigens, autoantibodies to, in cold idiotypic targets of, 245
Rous, Peyton, 17, 18f agglutinin syndrome, 245–246, Ii blood group, 246–249, 246t, 251t
Rubella, AIHA in, 106, 107t 251t immunoglobulin G, 245
Runt disease, 340 Sickle cell(s) Ju antigen, 253
Rx antigen, autoantibodies to, 239 irreversible, 326–327 Kell system, 236–237
in peripheral blood film, 53, 55f Kidd system, 237–238
reversible, 326–327 Li antigen, 250–251, 251t
Sickle cell disease Lud antigen, 251, 251t
S antigen, antibodies to, after transfusions, AIHA in, after transfusions, 336–337, LW blood group, 231–234, 253
542 553–555, 554f M antigen, 236, 253
Sa antigen, autoantibodies to, 245, 251, 251t blood film in, in sickle cell disease, 55f Me antigen, 253
Saline solution, low ionic-strength (LISS), CFAC test in, 325–327, 325t mimicking, 239–242, 241t
for DAT, 329, 332 transfusions for. See also Sickle cell N antigen, 236, 253
Sarcoidosis, AIHA in, 113 hemolytic transfusion reaction not associated with blood group
Sc antigens, autoantibodies to, 239 syndrome. antigens, 243–245, 243t
Schizont antigen, antibodies to, 109 alloimmunization in, 542 in paroxysmal cold hemoglobinuria,
Schubothe, H., 25 passenger lymphocyte syndrome in, 254
Schwartz, S.O., 16, 17f 359 Pr antigen, 249–250, 251t, 252t
Secondary autoimmune hemolytic RBC substitutes in, 396 relative, 385, 385t
anemia(s), 61, 62t, 83–113, 441–444 Sickle cell hemolytic transfusion reaction Rh, 231–234, 232t, 289–290, 385, 385t
in antiphospholipid syndrome, 94, 96 syndrome, 547–555 Sa antigen, 251, 251t
in autoimmune lymphoproliferative AIHA in, 553–555, 554f SP1 antigen, 249
syndrome, 112, 112t characteristics of, 547–550, 548t, 549f in stored red blood cells, 243, 243t,
in carcinoma, 97, 97t, 98t crisis in, 555 253–254
in chronic lymphocytic leukemia, definition of, 547 Tj(a) antigen, 254
441–444, 442t, 443f hyperhemolysis in, 552 after transfusions, 338
classification of, 82–83, 83t incidence of, 547 in transfusions, 385–387, 385t, 386f
in collagen disorders, 96 mechanisms of, 552 Vo antigen, 250–251, 251t
in cytomegalovirus infections, 102–103 pain crisis in, 547–548, 555 in warm antibody AIHA. See Warm
in hepatitis, 107–108 in pediatric patients, 551–552 antibody autoimmune
in HIV infection, 103–104, 105t in pregnancy, 551 hemolytic anemia.
idiopathic, 82–83, 82t reticulocytopenia in, 549 in younger red blood cells, 244
incidence of, 82–83, 83t–84t serologic findings in, 549–550 Spectrin, autoantibodies to, 244
in infections, 99–110, 101t, 105t, 107t severe anemia in, 548–549, 549f Spherocytes
in infectious mononucleosis, 101–102, treatment of, 555 discovery of, 7–8, 8f
101t with undetectable antibodies, 465 formation of, 146, 146f
in lymphoproliferative disorders, 88–94, Sideroblast(s), after splenectomy, 416 in peripheral blood film, 53, 53f, 54f
89t, 90f, 91t, 92t, 93f Sideroblastic anemia, AIHA in, 113 Spherocytosis
in malaria, 108–109 Siderocytes, after splenectomy, 416 in cold agglutinin syndrome, 73
in Mycoplasma pneumoniae infections, Silica, in false-positive direct antiglobulin in hemolytic anemia, 53t
100–101 test, 204 Ig in, 322, 322t
in ovarian tumors, 83–84, 85t Sjögren’s syndrome, AIHA in, 113 lactic dehydrogenase levels in, 45, 46t
in parasitic diseases, 108–110 Skin in paroxysmal cold hemoglobinuria, 76
in parvovirus B19 infections, 106–107 corticosteroid effects on, 406t in warm antibody AIHA, 66
in primary immunodeficiency, 110–112, necrosis of, in cold agglutinin syndrome, Spleen
111t 72 early studies of, 14–15, 16f
in rubella, 106, 107t SLE. See Systemic lupus erythematosus. irradiation of, for warm antibody AIHA,
splenectomy for, 409, 409f, 409t Sleeping sickness, AIHA in, 109 416
in systemic lupus erythematosus, 94, 95t, Smallpox vaccination, warm antibody red blood cell destruction in, 140, 148–150
96 AIHA after, 98 removal of. See Splenectomy.
in thymoma, 96 Solid organ transplantation. See under transplantation of. See Transplantation,
treatment of Transplantation. solid organ.
guidelines for, 448 SP antigens, as early name for Pr antigens, Splenectomy
options for, 402t 249 for AIHA, 322, 324
in ulcerative colitis, 84, 85t–89t Specificities DAT-negative, 334
after vaccination, 97–99 of alloantibodies, bystander immune in pediatric patients, 343
in varicella, 104, 106 hemolysis and, 364 in pregnancy, 351
Senescent cell antigen, autoantibodies to, of autoantibodies, 231–260 for cold agglutinin syndrome, 439–440,
243 ABO group, 238–239, 239t, 240t, 440t, 448
Septic shock, after splenectomy, 412 251–253 early studies of, 14–15, 16f
Index 589

Splenectomy (continued) Tazobactam, DIIHA from, 282–283 Transfusion(s) (continued)


Ig after, 322, 322t Teniposide, DIIHA from, 271 for AIHA (continued)
for paroxysmal cold hemoglobinuria, 441 Teratogenesis, of immunosuppressive autologous blood for, 396–397
in ulcerative colitis, AIHA remission therapy, 418 chronic stable, 377
after, 88 Teratoma, ovarian, AIHA in, 83–84, 85t compatibility testing in
for warm antibody AIHA, 67, 407–416, 447 Thalassemia(s) cold-antibody, 389–391, 390t
abnormal laboratory tests after, 415–416 AIHA in, 113 in vivo, 394–395
adverse effects of, 411–415, 413t, 414t, rituximab for, 422–423 warm-antibody, 378–389, 379t,
415f stem cell transplantation for, 431 381t–385t, 386f, 387t, 388t
clinical response in, 408–409, 408t, 409f, after transfusions, 337 complications of, 392–394, 393f
409t CFAC test in, 325–327, 325t fulminant, 377
indications for, 407 parvovirus B19 infections in, 107 in HIV infection, 104
long-term results of, 409–410 transfusions for indications for, 375
mechanisms of response to, 410 alloimmunization in, 542 leukocyte-reduced RBCs for, 396
patient selection for, 410–411, 411f hemolytic reactions after, 552 optimal volume in, 391–392
prediction of responses to, 410–411, Thioguanine, for warm antibody AIHA, 416 in pediatric patients, 343, 344
411f Thrombocytopenia rapidly progressing, 376, 377, 377t
technique for, 407–408 in AIHA reluctance to, 376
Spleno-hepatic index, in splenectomy in parvovirus B19 infections, 107 risks of, 378
results evaluation, 411, 411f in primary immunodeficiency diseases, stable, 376–377
Splenomegaly 110–111 substitutes for, 395–396
in AIHA alloimmune warm blood in, 395
in HIV infection, 104 after hematopoietic cell washed RBCs for, 396
in leukemia, 89 transplantation, 491–492 alloantibody transfer in, direct
in ovarian tumors, 85t after solid organ transplantation, 506 antiglobulin test in, 204
in autoimmune lymphoproliferative autoimmune, after hematopoietic cell autoantibody development after,
syndrome, 112 transplantation, 491 335–340, 335t
in infectious mononucleosis, 101 in autoimmune lymphoproliferative animal studies of, 335–336
in Mycoplasma pneumoniae infections, 100 syndrome, 112 human data on, 336–337
in warm antibody AIHA, 64 in hemolytic transfusion reactions, 557 immunological data on, 338–339
Stats, D., 25 immune, after hematopoietic cell in multiple transfusions, 337–338
Stem cell transplantation, for warm transplantation, 490–491 source of, 339–340
antibody AIHA, 431–432 in mixed AIHA, 81 case reports of, 392–394, 393f
Stomatocytes, 54 in paroxysmal cold hemoglobinuria, 76 for cold agglutinin syndrome, 448
Streptococcus pneumoniae infections after transfusion, 360 controversies over, 28–29
AIHA in, 110 in warm antibody AIHA, 66–68 for delayed hemolysis, after
after splenectomy, 412–413 Thrombocytopenic purpura hematopoietic cell
vaccines for, 413 idiopathic, after solid organ transplantation, 480, 480f
Streptomycin, DIIHA from, 270 transplantation, 506 for DIIHA, 291
Stress (shift) reticulocytes, 36, 39 thrombotic errors in, reporting of, 560–563
Stroke, from intravenous Ig, 429 AIHA in, 113 exchange
Super Coombs tests, 332–333, 332t, 333t after hematopoietic cell for HDFN, 532–533, 536
Syphilis, paroxysmal cold hemoglobinuria transplantation, 497–500, 498t of red blood cells, before hematopoietic
in, 74, 77, 78 peripheral blood film in, 56f cell transplantation, 469
Systemic lupus erythematosus in warm antibody AIHA, 67–68 fetal, for HDFN, 524, 532–533
AIHA in, 94, 95t, 96 Thrombocytosis granulocyte, alloantibody transfer in, 567
treatment of, 421, 444–445, 445f in paroxysmal cold hemoglobinuria, 76 guidelines for, for error prevention,
anticardiolipin antibodies in, 245 after splenectomy, 414–415, 415f 561–563
danazol for, 424 in warm antibody AIHA, 66–67 haptoglobin levels after, 44–45
differential diagnosis of, 177 Thromboembolism for HDFN, 532–533, 536
from fetal cell persistence, 340 in AIHA, in HIV infection, 104 before hematopoietic cell transplantation,
stem cell transplantation for, 432 from intravenous Ig, 429 blood products for, 480–481,
after splenectomy, 415 481f
in warm antibody AIHA, 64–66, 65f historical aspects of, 26, 27f
Thrombotic thrombocytopenic purpura irradiated products for, in hematopoietic
T lymphocytes AIHA in, 113 cell transplantation, 481, 481f
depletion of, for transplantation, after hematopoietic cell transplantation, least incompatible units in, 386–387
passenger lymphocyte 497–500, 498t multiple
syndrome and, 467 peripheral blood film in, 56f alloantibody tests for, 387–388
double-negative, in autoimmune Thymectomy, for warm antibody AIHA, alloimmunization in, 542
lymphoproliferative syndrome, 433, 433t, 434t complications of, 392–393, 393f
112 Thymoma, AIHA with, 96 for passenger lymphocyte syndrome,
in red blood cell destruction, 158 Thyroid disease, AIHA in, 113 470, 471t, 504, 504t
after transfusions, 338–339 Ticarcillin, DIIHA from, 286 patient identification for, 562–563
Tacrolimus (FK506) Ticarcillin/clavulanate, DIIHA from, 282 platelet
for graft-versus-host disease prevention, Tj antigens, autoantibodies to, in alloantibody transfer in, 567
passenger lymphocyte paroxysmal cold hemoglobinuria, hemolysis after, 494–495
syndrome and, 468 254 volume reduction in, 469
for solid organ transplantation, Transfusion(s). See also Transfusion positive DAT results after, 207
hemolysis related to, 502 reactions, hemolytic for pure red cell aplasia, after
Target cells for AIHA, 375–400 hematopoietic cell
in peripheral blood film, 53, 55f acute onset, 376 transplantation, 479–480
after splenectomy, 416 assessment of need for, 375–376 purpura after, 360
590 Index

Transfusion(s) (continued) Transplantation (continued) Warm antibody autoimmune hemolytic


requirements for solid organ (continued) anemia, 54f, 62–71
in hemolysis assessment, 46–48, 48t idiopathic thrombocytopenic purpura age distribution in, 63, 63f
normal, 47, 48t after, 506 antibody specificity in, 231–234, 232t
in passenger lymphocyte syndrome, immune hemolytic anemia after, ABO system, 238–239
463, 464t 504–505, 505f glycophorins and, 234–236, 235t
for sickle cell disease immune neutropenia after, 507 Hh system, 238–239
alloimmunization in, 542 microchimerism in, 507 Kell system, 236–237
passenger lymphocyte syndrome in, passenger lymphocyte syndrome after, mimicking, 240, 241t
359 500–504, 501t, 504t in stored red blood cells, 243, 253–254
testing before, positive DAT results in, Triethylene melamine, for warm antibody autoagglutination in, 168
207 AIHA, 416 in autoimmune lymphoproliferative
for thalassemia, alloimmunization in, 542 Trosier, J., 13 syndrome, 112
for warm antibody AIHA, 447 Trypanosomiasis, African, AIHA in, 109 blood picture in, 64t, 66–68
Transfusion reactions, hemolytic Tumor(s), ovarian, AIHA in, 83–84, 85t bone marrow findings in, 68
ABO-incompatible, 558–559 Tumor lysis syndrome, from rituximab, 423 case reports of, with negative DAT,
AIHA as, 335–340, 335t Tumor necrosis factor alpha, in hemolytic 323–326
animal studies of, 335–336 transfusion reactions, 556 clinical manifestations of, 63–64, 64t
bystander immune hemolysis in, 361 cold agglutinin syndrome with. See
versus delayed hemolytic transfusion Mixed autoimmune hemolytic
reactions, 356–357 anemia.
human data on, 336–337 U blood group, autoantibodies to, in cytomegalovirus infections, 103
immunological data on, 338–339 glycophorins and, 234, 235t versus delayed hemolytic transfusion
in multiple transfusions, 337–338 Ulcerative colitis, secondary AIHA in, 84, reaction, 356
source of, 339–340 85t–89t direct antiglobulin test in, 172t, 174, 176,
antibodies causing, 543–544, 543t, 544t Ultrasonography, in HDFN, 524 176t
case reports of, 558–559 Umbilical blood sampling, in HDFN, 524–526 after hematopoietic cell transplantation,
clinical features of, 556, 556t, 557f Umbilical cord stem cell transplantation, for 485, 486t
in coagulation factor transfusions, 567 Evans’ syndrome, 487 in HIV infection, 104
definition of, 541 Urine, hemoglobin in. See Hemoglobinuria. immunoglobulin A in, with negative
delayed. See Delayed hemolytic Urobilinogen, measurement of, 43 DAT, 330–331
transfusion reactions. immunoglobulin G in, 178–180, 179t
differential diagnosis of, 168 immunoglobulin M in, 180–182, 181t
direct antiglobulin test in, 204 with negative DAT, 331–332
disseminated intravascular coagulation Vaccination immunopancytopenia in, 68–69
with, 392, 557, 558 AIHA after, 97–99 incidence of, 62, 63t
due to antibodies not detectable by Haemophilus influenzae, 413 with negative DAT, 319–320
routine procedures, 563–567 influenza, 414 laboratory tests for, 25–26, 66–68, 169,
fatalities due to, 559–560 Streptococcus pneumoniae, 413 170t, 178–180, 178f, 179t,
in granulocyte transfusions, 567 van Leeuwenhoek, Antoni, 2, 3f 225t–227t
in group O transfusions, 566 Vancomycin, prophylactic, after negative DAT in, 332–333, 332t, 333t
historical aspects of, 26–28 splenectomy, 414t in lymphoma, 446
human leukocyte antigen antibodies in, Vanlair, C.F., 5, 7 misdiagnosis of, with negative DAT, 325
565 Varicella, AIHA in, 104, 106 negative DAT in, 323t
immediate, definition of, 541 Vel antigens, autoantibodies to, 239 case reports of, 323–326
incidence of, 541–545 Venous thromboembolism, in warm IgA in, 330–331
kidney failure with, 557–558 antibody AIHA, 64–66, 65f IgM in, 331–332
laboratory tests for, 556, 556t, 557f, Vinca alkaloids, for warm antibody AIHA, incidence of, 319–320
567–568 417 laboratory tests for, 332–333, 332t, 333t
non-ABO alloantibody passive transfer Vincent, C., 13 misdiagnosis of, 325
in, 566–567 Vincristine in pediatric patients, 103, 324t, 341–344,
nonhumoral mechanisms in, 565–566 for AIHA, in chronic lymphocytic 341t, 342f, 433
passively transfused alloantibodies in, leukemia, 444 physical signs in, 64–66, 64t, 65f, 69f
566 for warm antibody AIHA, 417 in pregnancy, 345
pathophysiology of, 555–558, 556t, 557f Viral infections. See also specific infections. prognosis for, 70–71
in plasma transfusions, 567 AIHA in, plasma exchange for, 430 recovery from, 71
in platelet transfusions, 494–495, 567 cold agglutinin syndrome in, 113 reticulocytes in, 69–70, 69f, 69t
reporting of incidents, 560–563 from immunosuppressive therapy, 418 secondary, 64
severe, bystander immune hemolysis in, paroxysmal cold hemoglobinuria in, 74–75 serology of, 178–180, 178f, 179t
359–360 transmission of, in intravenous Ig, 428 sex distribution in, 63
in sickle cell disease, 547–555, 548t, 549f, Visceral leishmaniasis, AIHA in, 109 in SLE, 444–445
554f Vitamin D supplements, with corticosteroid survival in, 70–71
treatment of, 558 therapy, 406 after transfusions, 337
types of, 541 Vo antigen, autoantibodies to, 250–251, 251t transfusions for, compatibility testing for,
Transplantation Vogel, J., 4 378–389, 379t, 381t–385t, 386f,
hematopoietic. See Hematopoietic cell Volume overload, in transfusions, 392 387t, 388t
transplantation; specific types. treatment of, 401–433. See also
solid organ Immunosuppressive therapy,
AIHA after, 506 for warm antibody AIHA.
alloantibodies of recipient origin after, Waldenström’s macroglobulinemia corticosteroids in, 401–407, 403t, 405t,
505–506 cold agglutinin syndrome in, 113–114, 406t
alloimmune thrombocytopenia after, 113t, 114t, 189–190 guidelines for, 447
506 mixed AIHA with, 81 options for, 402t
Index 591

Warm antibody autoimmune hemolytic Wasserman, L.R., 25 Yersinia enterocolitica infections, AIHA in,
anemia (continued) White blood cell count 110
treatment of (continued) in corticosteroid therapy, 408 Yt antigens, antibodies to, in hemolytic
plasma exchange in, 429–431 after splenectomy, 415 transfusion reactions, 543
in SLE, 444–445 in warm antibody AIHA, 66
splenectomy in, 408–416, 408t, 409f, Widal, Fernand, 13–14
409t, 411f, 413t, 414t, 415f Wiener, A.S., 18–19, 20, 25
splenic irradiation in, 416 Wiskott-Aldrich syndrome, AIHA in, 111, Zymosan, in complement activation, 138
stem cell transplantation in, 431–432 111t ZZAP reagent
thymectomy in, 433, 433t Witebsky, E., 23 in allogeneic adsorption test, 384–385
after vaccination, 98–99 Wr antigen, autoantibodies to, 234–236 in warm autoadsorption technique,
Warm autoadsorption technique, for 380–383
alloantibody detection, 382–383,
383t
Warm-reactive autoantibodies, 61, 62t Xg antigen, autoantibodies to, 239

You might also like