You are on page 1of 7

Review

How to Manage Neutropenia in Multiple Myeloma


Antonio Palumbo,1 Jon Bladé,2 Mario Boccadoro,1 Carmela Palladino,1
Faith Davies,3 Meletios Dimopoulos,4 Anna Dmoszynska,5 Hermann Einsele,6
Philippe Moreau,7 Orhan Sezer,8 Andrew Spencer,9 Pieter Sonneveld,10
Jesus San Miguel11

Abstract
Neutropenia is a hematologic adverse event characterized by an absolute neutrophil count (ANC) lower than 1500
cells/mL. This reduction may be due to decreased neutrophil production, accelerated use, a shift in compartments
of neutrophils, or a combination of these factors. Neutropenia is often associated with infections, which are major
causes of morbidity and mortality in patients with cancer. In patients with multiple myeloma, the novel agents
thalidomide, lenalidomide, and bortezomib have improved outcome, but chemotherapy-related neutropenia should
be carefully considered. Chemotherapy-related high-risk factors for severe neutropenia include regimens with an
expected neutropenia rate of ⬎ 50%, such as the 3-drug combinations including lenalidomide plus alkylating agents
or doxorubicin, whereas low-risk regimens include combinations of the novel agents with dexamethasone alone.
Patient characteristics, disease stage, type of current and previous treatment, and ANC ⬍ 1000 cells/mL at baseline
are additional factors that define the risk of severe neutropenia.
Granulocyte-colony stimulating factor (G-CSF) should be used to manage chemotherapy-related neutropenia so
that patients may stay on treatment for a longer time and benefit from it. Primary G-CSF prophylaxis should be used
when high-risk regimens are administered or when low/intermediate-risk regimens are used and additional risk
factors are present. Reactive G-CSF treatment is indicated when patients undergoing low-risk chemotherapy
experience grade 3/4 neutropenia. If ANC restores to ⬎ 1000 cells/mL, therapy can be resumed with no dose
modifications. In case of persistence of severe neutropenia, treatment should be delayed until ANC reaches ⬎ 1000
cells/mL, and dose reductions are necessary.

Clinical Lymphoma, Myeloma & Leukemia, Vol. 12, No. 1, 5-11 © 2012 Elsevier Inc. All rights reserved.
Keywords: Absolute neutrophil count, Granulocyte colony-stimulating factor, Multiple myeloma,
Myelosuppression, Neutropenia

Definition and Classification of trophils originate from pluripotential stem cells in the bone mar-
Neutropenia row.1 Two hematopoietic growth factors, namely granulocyte colo-
Neutropenia is a blood disorder related to neutrophilic white cells, ny-stimulating factor (G-CSF) and granulocyte-macrophage colony-
which are essential to prevent bacterial and fungal infections. Neu- stimulating factor, regulate the production and deployment of
neutrophils.2 Healthy adults have an absolute neutrophil count
(ANC) of about 1500-7000 cells/mL (1.5-7.0 ⫻ 109/L). According
1
Myeloma Unit, Division of Hematology, University of Torino, AOU San Giovanni to the National Cancer Institute Common Terminology Criteria for
Battista, Torino, Italy
2
Department of Hematology, Hospital Clinic, Barcelona, Spain Adverse Events (NCI CTCAE) V3.0, it is classified as mild (ANC
3
Institute of Cancer Research and Royal Marsden Hospital, London, United
Kingdom
4
Department of Clinical Therapeutics, University of Athens School of Medicine, 10
Department of Hematology, University Hospital Rotterdam, Rotterdam,
Alexandra General Hospital, Athens, Greece Netherlands
5
Department of Haematooncology and Bone Marrow Transplantation, Medical 11
Servicio de Hematología, Hospital Universitario de Salamanca, and CIC, IBMCC
University of Lublin, Lublin, Poland (USAL-CSIC), Salamanca, Spain
6
Department of Internal Medicine II, University Hospital Würzburg, Würzburg,
Germany Submitted: Aug 4, 2011; Revised: Oct 14, 2011; Accepted: Nov 9, 2011
7
Hematology Department, University Hospital Hôtel-Dieu, Nantes, France
8
Department of Hematology and Oncology, University Medical Center, Hamburg, Address for correspondence: Antonio Palumbo, Myeloma Unit, Division of
Germany Hematology, University of Torino, AOU San Giovanni Battista, Torino, Italy
9
Malignant Hematology and Stem Cell Transplantation Service, Alfred Hospital, Fax: ⫹39 01 1696 3737; e-mail contact: appalumbo@yahoo.com
Melbourne, Australia

2152-2650/$ - see frontmatter © 2012 Elsevier Inc. All rights reserved.


doi: 10.1016/j.clml.2011.11.001 Clinical Lymphoma, Myeloma & Leukemia February 2012 5
How to Manage Neutropenia in Multiple Myeloma
tively.12,13 These drugs, particularly when used in association with
Table 1 Neutropenia Severity According to the National
Cancer Institute Common Toxicity Criteria standard chemotherapy in elderly and transplant-ineligible patients,
may cause severe myelosuppression.14 Since severe neutropenia is a
Grade ANC (cells/mL) relevant side effect of lenalidomide, a group of experts has provided
0 Within normal limits some clinical guidance for the management of cytopenia. In general,
the blood count should be monitored on a biweekly basis during
1 ⱖ 1500 to ⬍ 2000
lenalidomide treatment, but weekly monitoring is recommended in
2 ⱖ 1000 to ⬍ 1500
patients with cytopenia at baseline. In case of cytopenia, G-CSF or
3 ⱖ 500 to ⬍ 1000 erythropoietin stimulating agents may be necessary, and in some
4 ⬍ 500 cases dose reductions or interruptions may be needed.15 Routine
antibiotic use is recommended for all patients on initiation of lena-
Abbreviation: ANC ⫽ absolute neutrophil count.
lidomide treatment.16
This article focuses on neutropenia in multiple myeloma and aims
1000-1500 cells/mm3), moderate (ANC 500-1000 cells/mm3), and
to provide clinicians with a simple algorithm to deal with this side
severe (ANC ⬍ 500 cells/mm3) (Table 1).
effect. This article is based on the consensus achieved among the
Febrile neutropenia is classified as an ANC lower than 1000 cells/
authors. Amgen has provided support to write the guidelines but did
mm3 (⬍ 1.0 ⫻ 109/L) and with fever higher than 38.3°C (101°F) or
not provide any comment on the content, data analysis, or
a sustained temperature of ⱖ 38°C (100.4°F) for more than 1 hour
interpretation.
(NCI CTCAE V4.0).
Severe neutropenia can lead to serious problems and requires im-
mediate and careful care because the patient could potentially ac-
Risk Factor Assessment
quire a bacterial, fungal, or mixed infections at any time. Symptoms Low ANC before starting treatment is the primary risk factor for
depend on the level and grade of neutropenia. The lower the neutro- the development of severe neutropenia and neutropenic complica-
phil count, the greater the grade of neutropenia (Table 1) and con- tions after chemotherapy.17–19 Additional risk factors can be identi-
sequently the higher the risk of infection. This risk normally in- fied. They can be classified as patient-specific, cancer-specific, or
creases if the neutrophil count remains less than the normal level for treatment-specific (Table 2). Patient-specific risk factors are female
more than 3 days. Types of infection include otitis media, tonsillitis, sex, a small body surface area (BSA) (⬍ 2 m2) and age ⬎ 65 years.
sore throat, mouth ulcers, gum infection, and skin abscesses. Any These factors represent a major risk for chemotherapy-related toxic-
fever (body temperature ⬎ 38.5°C or 101.3°F) must be taken very ity in oncology and are associated with severe myelosuppression be-
seriously into account and the physician should be informed. Signs of cause of high systemic absorption of drugs resulting in increased
infection, such as swelling and warmth, are not usually present in toxicity. Comorbidities such as diabetes mellitus and cardiovascular
neutropenia-associated infections.3 disease as well as the presence of open wounds and altered hepatic or
renal function, which are typical of the elderly, poor nutritional sta-
Neutropenia in Multiple Myeloma tus with low serum albumin levels, and low performance status
Multiple myeloma is the second most common hematologic ma- highly increase the risk of neutropenic complications.17,18,20 –22
lignancy. It accounts for 1% of all cancers and 13% of hematologic Cancer-specific risk factors for neutropenia include type and status
neoplasms; it is typically seen in the elderly, and the median age at of cancer. The more the abnormal process of growth involves the
diagnosis is 70 years,4 – 6 Although the disease remains incurable, the bone marrow, the higher the severity and grade of cytopenia in pe-
introduction of novel agents such as the immunomodulatory drugs ripheral blood, and the slower the recovery after chemotherapy. Pa-
thalidomide and lenalidomide and the proteasome inhibitor bort- tients who have already received chemotherapy and have aggressive
ezomib, has markedly changed the treatment paradigm of myeloma or relapsed/refractory disease are at higher risk for the development
and considerably improved outcomes.7 The efficacy of these drugs of grade 3/4 neutropenia.17,19 Treatment-specific risk factors in-
should always be balanced against their toxicity profile. Therapy- clude previous exposure to chemotherapy or radiation therapy,
related toxicities may in fact negatively affect outcome and lead to which contributes to cytopenia and neutropenia. Of note, all patients
early treatment discontinuation. Neutropenia is a hematologic side who are treated with chemotherapy are at risk for neutropenic com-
effect commonly associated with chemotherapy.8,9 plications.17,19 The type of chemotherapeutic regimen is one of the
In myeloma patients receiving new drugs, constant monitoring primary determinants of the risk of neutropenia in multiple my-
may predict occurrence of neutropenia. Neutropenia can also be eloma. Some chemotherapeutic regimens are more myelotoxic than
managed with appropriate supportive case management, dose adjust- others. High-dose alkylating agents and novel agent– containing
ment, and growth factor support. In the era of novel agents, neutro- therapies for multiple myeloma can cause myelosuppression, espe-
penia is less common, especially with thalidomide and bortezomib, cially when used in combination.23
unless they are combined with conventional agents, such as alkylat- Based on the risk of chemotherapy-induced neutropenia in pa-
ing agents. By contrast, lenalidomide-induced neutropenia should tients with multiple myeloma, 4 categories of risk regimens can be
always be considered because neutropenia is one of the most frequent identified: low, low/intermediate, intermediate, and high (Table 3).
side effects of lenalidomide. The reported incidence of grade 3/4 Overall, in patients who receive chemotherapy for the first time and
neutropenia with lenalidomide is approximately 35%,10,11 and it is have no other risk factors, low-risk regimens include novel agents in
about 11% and 1% with bortezomib and thalidomide, respec- association with dexamethasone, such as lenalidomide in association

6 Clinical Lymphoma, Myeloma & Leukemia February 2012


Antonio Palumbo et al
Table 2 Risk Factors for Neutropenia

Patient-Related Cancer-Related Treatment-Related


Age > 65 years Heavy marrow infiltration History of severe neutropenia with previous chemotherapy
Comorbidities
Female Sex Advanced/uncontrolled disease High-risk chemotherapy
Poor Nutritional Status (low albumin)
Decreased Immune Function Relapsed/refractory disease Extensive previous chemotherapy
Low Performance Status
Small Body Surface Area < 2 m2 Preexisting neutropenia Concurrent or previous radiation therapy

Table 3 Neutropenia Risk Associated with Each Regimen few years, the mortality rate due to febrile neutropenia has decreased
steadily, but it remains significant. In some hematologic malignan-
Grade 3/4 < 50% Grade 3/4 > 50% cies, overall mortality from febrile neutropenia reaches 14%, whereas
Low Risk
Low/Intermediate Intermediate
High Risk for multiple myeloma it is around 8%.34 Neutropenia alters the
Risk Risk host’s inflammatory response, making it difficult to detect the usual
RD MPT PAD RAD signs and symptoms of infection. A careful history and a detailed and
VD CTD CVD MPR repeated physical examination are thus necessary. In patients with
TD MPV CRD
central venous catheters, at least 2 sets of blood cultures from the
catheters and from a peripheral site, as well as cultures from other
VTD
sites (eg, urine, stool, throat), should be obtained before starting
VRD empirical antibiotic therapy. Cultures should be repeated daily while
Abbreviations: CRD ⫽ cyclophosphamide-lenalidomide-dexamethasone; CTD ⫽ cyclophosph- patients remain febrile. Chest radiography followed by imaging tech-
amide-thalidomide-dexamethasone; CVD ⫽ cyclophosphamide-bortezomib-dexamethasone; niques such as high-resolution computed tomography (CT) is essen-
MPR ⫽ melphalan-prednisone-lenalidomide; MPT ⫽ melphalan-prednisone-thalidomide;
MPV ⫽ melphalan-prednisone-bortezomib; PAD ⫽ bortezomib-Adriamycin-dexamethasone; tial to detect pulmonary lesions. CT, magnetic resonance imaging,
RAD ⫽ lenalidomide-Adriamycin-dexamethasone; RD ⫽ lenalidomide-dexamethasone; ultrasonography, and radionuclide imaging of any potential site of
TD ⫽ thalidomide-dexamethasone; VD ⫽ bortezomib-dexamethasone; VRD ⫽ bortezomib-
lenalidomide-dexamethasone; VTD ⫽ bortezomib-thalidomide-dexamethasone. infection must be performed. Invasive procedures such as broncho-
scopic examination and lung, liver, or skin biopsy are also useful to
detect infections.
with dexamethasone (RD),24 bortezomib plus dexamethasone It is hard to predict the risk of complications related to febrile neutro-
(VD),25 and thalidomide plus dexamethasone (TD).26 Low/inter- penia. Differentiating between high- and low-risk patients has a signifi-
mediate-risk regimens include thalidomide plus melphalan and cant impact on the patients’ quality of life and outcome, as well as on
prednisone (MPT)27 and thalidomide plus cyclophosphamide and medical costs for hospitalization.8,35 The degree of neutropenia and its
dexamethasone (CTD). Intermediate-risk regimens include bort- duration are important aspects to consider. Patients who are expected to
ezomib-doxorubicin-dexamethasone (PAD),28 cyclophosphamide- recover their granulocyte counts in ⬍ 1 week are generally considered at
bortezomib-dexamethasone (CVD), bortezomib-melphalan-predni- low risk of complications after onset of fever. High-risk patients experi-
sone (VMP),29 and the combinations of bortezomib with ence prolonged neutropenia lasting ⬎ 7 days.36
dexamethasone plus either thalidomide (VTD)30 or lenalidomide An internationally validated scoring system to classify patients
(VRD). High-risk regimens include the 3-drug combinations con- with cancer according to the risk of febrile neutropenia has been
taining lenalidomide plus doxorubicin-dexamethasone (RAD),31 developed by the Multinational Association of Supportive Care in
melphalan-prednisone (MPR), cyclophosphamide-dexamethasone Cancer (MASCC) and is shown in Table 4. This numerical risk score
(CRD), or melphalan-prednisone (MPR),32 in which the expected takes into account different features that are likely to contribute to a
incidence of neutropenia is higher than 50%. favorable outcome. A high global score indicates a high probability of
Besides the neutropenia risk associated with the specific regimen, fever resolution with no serious complications. A MASCC risk score
patient and disease characteristics remain fundamental to assess the ⱖ 21 identifies low-risk patients with a positive predictive value of
final risk category. In particular, patients with preexisting cytopenia 91%, a specificity of 68%, and a sensitivity of 71%.37
and high tumor burden in the bone marrow are at higher risk of Based on this score index, high-risk patients have a global risk
severe neutropenia.14,33 score ⬍ 21. G-CSF is not routinely indicated as an adjunct to anti-
biotic therapy in uncomplicated fever and neutropenia. However the
Febrile Neutropenia use of G-CSF should be considered in patients at high risk for com-
Febrile neutropenia is defined as a single oral temperature reading plications, or when prognostic factors are predictive of poor clinical
higher than 38.3°C (101°F) or a sustained (more than 1 hour) tem- outcome, including expected prolonged (⬎ 10 days) and profound
perature of ⱖ 38°C (100.4°F) in a patient with an ANC lower than (ANC ⬍ 1000 cells/mL) neutropenia, age ⬎ 65 years, uncontrolled
1000 cells/mm3 (⬍ 1.0 ⫻ 109/L) (NCI CTCAE V4.0). In the past primary disease, pneumonia, hypotension and multiorgan dysfunc-

Clinical Lymphoma, Myeloma & Leukemia February 2012 7


How to Manage Neutropenia in Multiple Myeloma
Clinical data support the use of primary G-CSF prophylaxis in
Table 4 Multinational Association of Supportive Care in
Cancer Index patients receiving chemotherapy regimens with an expected febrile
neutropenia rate higher than 20%, as well as in those with a febrile
Variables Score neutropenia rate of 10%-20% when at least 1 patient-related risk
Burden of Illness factor for febrile neutropenia is present.20,44
A different approach is recommended when severe neutropenia
No or mild symptoms 5
occurs in a patient with multiple myeloma who has been treated
Moderate symptoms 3
with chemotherapy with a low or low/intermediate risk of severe
Severe symptoms 0 neutropenia and who does not present with any other risk factors.
No Hypotension (Systolic blood pressure In these patients, reactive G-CSF prophylaxis should be used, that
5
> 90 mm Hg)
is to say prophylaxis is administered only after the occurrence of
No Chronic Obstructive Pulmonary Disease 4 the adverse event. If neutropenia resolves and ANC restores to ⬎
Solid Tumor/Lymphoma With No Previous
4 1000 cells/mL after G-CSF administration, therapy can be re-
Fungal Infection sumed and no dose modifications are needed (Figure 1). On the
No Dehydration 3 contrary, if ANC remains lower than 1000 cells/mL, treatment
Outpatient Status (at Onset of Fever) 3 should be delayed until ANC recovers to ⬎ 1000 cells/mL, and
Age < 60 Years 2 dose reductions are necessary (Table 5). If the patient experiences
another severe neutropenic event after the first dose reduction,
A global score ⱖ 21 denotes low risk of complications. Points attributed to the variable “burden treatment should be suspended again. It can be resumed when the
of illness” are not cumulative. The maximum theoretical score is therefore 26.
ANC rises to ⬎ 1000 cells/mL and treatment can be resumed
with further dose reductions (Table 5). Finally, in patients with
high tumor burden in the bone marrow, full-dose chemotherapy
tion (sepsis syndrome), invasive fungal infection, and hospitalization
is recommended; consequent myelosuppression is not to be
at the time of development of fever.20,38 – 42
treated with G-CSF administration, but monitoring until recov-
Recommendations for the Management of Novel ery is suggested, unless complications occur (fever, infection).
Therapy–related Neutropenia In Multiple Myeloma
In patients with multiple myeloma who are treated with novel Dose and Timing of G-CSF
agent– containing chemotherapy, the risk of severe neutropenia and Since the kinetics and recovery of bone marrow function vary
neutropenic complications should be carefully taken into account. between cytotoxic agents and regimens, it is difficult to provide gen-
Particular attention should be paid to febrile neutropenia, since it eral recommendations on dose and timing.
requires hospitalization, involves substantial costs, and has mortality G-CSF is contraindicated within 24 hours of chemotherapy, when
rates that are not negligible.43 stimulation of progenitor cells in the presence of cytotoxic agents
To limit and manage neutropenia and the related complications, sim- may worsen the myelotoxicity of the regimen. For cytotoxic drugs
ple guidelines for physicians to appropriately use G-CSF are needed. with a long half-life, a longer interval may be required to avoid in-
G-CSF is a glycoprotein that stimulates survival, proliferation, differen- creased myelotoxicity.39,42
tiation, and function of neutrophil granulocyte progenitor cells and ma- Subcutaneous and intravenous G-CSF have equivalent potency, but
ture neutrophils. The two forms of recombinant human G-CSF in clin- subcutaneous injection is generally recommended, as it is suitable for
ical use (filgrastim and lenograstim) are potent stimulants of neutrophil outpatients.45
granulopoiesis and have demonstrated efficacy in preventing infectious Lenograstim 150 ␮g/m2 once daily by subcutaneous injection
complications of some neutropenic states.41 (vial/syringe size 105 ␮g and 263 ␮g) is commonly used in practice
Two major strategies for the use of G-CSF are recommended, as in a daily dose of 263 ␮g (patients with BSA ⬎ 1.8 m2 may be given
either primary or reactive prophylaxis. Primary G-CSF prophylaxis is an additional 105 ␮g daily, either initially or if they have an inade-
recommended when using chemotherapy regimens associated with a quate response to the standard daily dose).20
high risk of the development of grade 3/4 neutropenia, when the Filgrastim 5 ␮g/kg is used once daily by subcutaneous injection
expected neutropenia rate is ⬎ 50%. Thus G-CSF prophylaxis is (vial/syringe size 300 ␮g and 480 ␮g). It is common practice for a
recommended with 3-drug regimens containing lenalidomide, such daily dose of 300 ␮g to be given; however patients who weigh ⬎ 90
as MPR, CRD, and RAD. In addition, primary G-CSF prophylaxis kg may be given the 480-␮g syringe either initially or if they have
should be used with low/intermediate-risk regimens with an ex- inadequate response to the standard dose.
pected severe neutropenia incidence ⬍ 50%, such as the 2-drug Lenograstim and filgrastim should be started not before 24 hours
regimens RD, VD, and TD and the 3-drug combinations MPT, and not later than 72 hours after cytotoxic treatment is completed. In
CTD, PAD, MPT, VMP, VTD, and VRD, only if 1 or more pa- this way the stimulus provided by G-CSF will be present at a time
tient- or cancer-related risk factors are also present and ANC is ⬍ when the bone marrow is regenerative and able to respond. G-CSF
1000 cells/mL at the start of chemotherapy (Table 3). Moreover it is injections should continue until ANC has recovered to at least 1000
strongly recommended that the dose be adjusted correctly based on cells/mL on 2 consecutive days.46
BSA when using chemotherapy regimens with a high risk for the Filgrastim is recommended to manage febrile neutropenia. It should
development of grade 3/4 neutropenia. be administered at the standard dose of 5 ␮g/kg daily, starting the day

8 Clinical Lymphoma, Myeloma & Leukemia February 2012


Antonio Palumbo et al
Figure 1 Algorithm for the Use of G-CSF in Patients with Myeloma Who are Receiving Chemotherapy

Grade 3-4 neutropenia observed

action
Add G-CFS and maintain doses

Does patient recover to ANC > 1000 ?

YES NO
action

action
Delay therapy until ANC ≥1000
Continue therapy at the same doses +
Resume treatment at reduced doses*

Abbreviations: ANC ⫽ absolute neutrophil count; G-CSF ⫽ granulocyte-colony stimulating factor. * Dose reductions are reported in Table 5.

Table 5 Suggested Dose Reductions When Grade 3/4 Events Do Not Resolve Despite G-CSF

Drug Full Dose Suggested Dose Reduction Further Reduction, if Needed


Melphalan 0.25 mg/kg days 1-4 for 4-6 weeks 0.18 mg/kg days 1-4 for 4-6 weeks 0.13 mg/kg days 1-4 for 4-6 weeks
Thalidomide 100 mg/day 50 mg/day 50 mg every other day
Lenalidomide 25 mg/day days 1-21 for 4 weeks 15 mg/day days 1-21 for 4 weeks 10 mg/day days 1-21 for 4 weeks
1.3 mg/m2 twice weekly days 1, 4, 8, 1.3 mg/m2 once weekly days 1, 8, 15, 22 for 5 1.0 mg/m2 once weekly days 1, 8, 15, 22 for
Bortezomib
11 for 3 weeks weeks 5 weeks
100 mg/day days 1-21 for 4 weeks or
50 mg/day days 1-21 for 4 weeks or 150 mg/m2/ 50 mg every other day days 1-21 for 4 weeks
Cyclophosphamide 300 mg/m2/day days 1, 8, 15 for 4
day days 1, 8, 15 for 4 weeks or 75 mg/m2/day days 1, 8, 15 for 4 weeks
weeks

after chemotherapy and finishing after 10-14 days, when the postnadir once in each chemotherapy cycle, in unique administration, 24 hours
neutrophil level has recovered to 1000 cells/mL. G-CSF is repeated with after completion of chemotherapy (Table 6).20,39,40 – 42,47
each cycle of chemotherapy.47
In patients receiving autologous bone marrow transplantation, the Conclusion
starting dose of G-CSF is 5 ␮g/kg daily, starting within 24-120 hours The combination of novel agents with conventional chemother-
after administration of high-dose therapy and tapering the dose accord- apy or dexamethasone has substantially changed the treatment par-
ing to neutrophil levels during the postnadir neutrophil recovery. adigm of patients with multiple myeloma, prolonging disease-free
The recommended dose of G-CSF for peripheral blood stem cell and overall survival. Treatment-related toxicities remain a main con-
mobilization is 10 ␮g/kg daily subcutaneously starting 4 days before cern in the era of new drugs as well. Neutropenia is a main concern
the first leukopheresis and ending after the last one. with lenalidomide, and caution is suggested when thalidomide and
Pegylated G-CSF (pegfilgrastim/pegG;) has recently been intro- bortezomib are used for the treatment of frail patients.14
duced. It is a novel G-CSF with double molecular weight and an A simple algorithm to manage neutropenia associated with novel
attenuated systemic clearance. It can be used to obtain sustainable therapies for multiple myeloma may help physicians to deal with this
G-CSF levels in particularly myelotoxic regimens at the dose of 6 mg adverse event appropriately. G-CSF prophylaxis is an optimal strat-

Clinical Lymphoma, Myeloma & Leukemia February 2012 9


How to Manage Neutropenia in Multiple Myeloma
Table 6 Recommendations for G-CSF Use

Growth Factor Setting Start Duration Dose


Myelotoxic 24-72 hours after administration of 5 ␮g/kg/day
Until ANC ⱖ 1000/mL
chemotherapy chemotherapy subcutaneously
Dose tapering according to
High-dose therapy and 24-120 hours after administration of 5 ␮g/kg/day
G-CSF (filgrastim) neutrophil levels during the post
ASCT rescue high-dose therapy subcutaneously
nadir neutrophil recovery
10 ␮g/kg/d
PBSC mobilization ⱖ 4 days before first leukopheresis Continue until last leukopheresis
subcutaneously
Pegylated G-CSF Myelotoxic Once per each chemotherapy
24 hours after end of chemotherapy 6 mg
(pegfilgrastim) chemotherapy cycle

Abbreviations: ANC ⫽ absolute neutrophil count; ASCT ⫽ autologous stem cell transplantation; G-CSF ⫽ granulocyte-colony stimulating factor; PBSC ⫽ peripheral blood stem cell.

egy to maintain chemotherapy at the desired dose intensity and min- search support from Celgene, Janssen-Cilag, and Onyx and has
imize delays. G-CSF prophylaxis is suggested with chemotherapy served on the advisory boards of Celgene, Janssen-Cilag, Millen-
regimens with a high risk of neutropenia. nium, and Onyx; Jesus San Miguel has served on the advisory
G-CSF should be used as a reactive treatment if severe neutropenia boards of Millennium, Celgene, and Johnson & Johnson.
occurs after chemotherapy in order to reduce the duration of neutro-
penia and related complications. If neutropenia does not resolve de- References
spite the use of G-CSF, treatment should be delayed until ANC 1. Metcalf D. Cellular hematopoiesis in the twentieth century. Semin Hematol 1999;
restores to ⬎ 1000 cells/mL. Treatment can then be resumed with 36:5-12.
2. Hannen M, Banning U, Bonig H, et al. Cytokine-mediated regulation of granulo-
appropriate dose reductions. This approach may reduce the risk of cyte colony-stimulating factor production. Scand J Immunol 1999; 50:461-8.
early treatment discontinuation for severe neutropenia, thus en- 3. Schwartzberg LS. Neutropenia: Etiology and pathogenesis. Clin Cornerstone; 2006;
8:S5-11.
abling patients to derive the highest benefit from the therapy. 4. Jemal A, Siegel R, Xu J, et al. Cancer statistics, 2010. CA Cancer J Clin 2010;
60:277-300.
5. Brenner H, Gondos A, Pulte D. Recent major improvement in long-term survival of
Acknowledgement younger patients with multiple myeloma. Blood 2008; 111:2521-6.
This study was supported by Amgen. The authors thank the edi- 6. Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple my-
eloma and the impact of novel therapies. Blood 2008; 111:2516-20.
torial assistant Giorgio Schirripa. 7. Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med 2004; 351:1860-73.
8. Klastersky J, Paesmans M, Rubenstein EB, et al. The Multinational Association for
Supportive Care in Cancer risk index: a multinational scoring system for identifying
Disclosures low-risk febrile neutropenic cancer patients. J Clin Oncol 2000; 18:3038-51.
Antonio Palumbo has received honoraria from Celgene, Jans- 9. Leong DC, Kinlay S, Ackland S, et al. Low-risk neutropenia in a medical oncology
unit. Aust N Z J Med 1997; 27:403-7.
sen-Cilag, Merck, and Amgen, and is on the advisory committees 10. Pérez Persona E, Mesa MG, García Sánchez PJ, et al. Lenalidomide treatment for
of Celgene and Janssen-Cilag; Jon Bladé has received honoraria patients with multiple myeloma: diagnosis and management of most frequent ad-
verse events. Adv Ther 2011;28:11-6.
from and has served on the advisory boards of Jansen-Cilag, Cel- 11. Palumbo A, Miguel JS, Sonneveld P, et al. Lenalidomide: a new therapy for multiple
gene, Novartis, and Pharmamar and has received research grants myeloma. Cancer Treat Rev 2008; 34:283-91.
12. San Miguel J, Bladé J, Boccadoro M, et al. A practical update on the use of bort-
from Jansen-Cilag and Celgene; Faith Davies has participated in ezomib in the management of multiple myeloma. Oncologist 2006; 11:51-61.
advisory boards and spoken at meetings for Celgene, Novartis, 13. Perri AJ 3rd, Hsu S. A review of thalidomide’s history and current dermatological
applications. Dermatol Online J 2003; 9:5
and Ortho-Biotech and has received travel support to attend 14. Palumbo A, Mateos MV, Bringhen S, et al. Practical management of adverse events
meetings from Celgene and Ortho Biotech; Meletios Dimopoulos in multiple myeloma: can therapy be attenuated in older patients? Blood Rev 2011;
25:181-91.
has received honoraria from Celgene and Ortho-Biotech; Mario 15. Sonneveld P, Dimopoulos M, San Miguel J, et al. Recommended management of
Boccadoro has received research support from and served on the cytopenia for len/dex in MM. Haematologica 2007; 92(s2):217: abstract PO-1122.
16. Attal M, Dimopoulos M, San Miguel J, et al. Management of non-haematological
consultancy and the scientific advisory boards of Celgene and toxicity of len/dex. Haematologica 2007;92(s2):217-8: abstract PO-1123.
Janssen-Cilag; Anna Dmoszynska has received honoraria from 17. Friese C. Chemotherapy induced neutropenia: important new data to guide nursing
assessment and management. Cancer Ther Supportive Care 2006;4:21-5.
Celgene and Janssen-Cilag and has served on the advisory com- 18. Aapro MS, Cameron DA, Pettengell R, et al. EORTC guidelines for the use of
mittees of Amgen and Mundipharma; Hermann Einsele has re- granulocyte-colony stimulating factor to reduce the incidence of chemotherapy-
induced febrile neutropenia in adult patients with lymphomas and solid tumours.
ceived research support from and has served on the advisory Eur J Cancer 2006; 42:2433-53.
boards of Celgene and Janssen-Cilag and has served on the speak- 19. Lyman GH, Lyman CH, Agboola O; ANC Study Group. Risk models for predict-
ing chemotherapy-induced neutropenia. Oncologist. 2005; 10: 427-37.
er’s bureaus of Celgene, Janssen-Cilag, and Novartis; Hermann 20. Smith T, Khatcheressian J, Lyman G, et al. 2006 update of recommendations for
Einsele has served on the advisory boards of Celgene, Novartis, the use of white blood cell growth factors: an evidence-based clinical practice guide-
lines. J Clin Oncol 2006; 24:3187-205.
and Janssen-Cilag; Orhan Sezer has received honoraria from Cel- 21. NCCN Clinical Practice Guidelines in Oncology Myeloid Growth Factors
gene, Janssen-Cilag, Amgen, and Novartis and has received re- (Version 1.2010). Available at: http://www.nccn.org/professionals/physician_gls/
f_guidelines.asp. Accessed November 21, 2011.
search funding from Janssen-Cilag and Novartis; Andrew Spencer 22. Crawford J, Dale DC, Lyman GH. Chemotherapy-induced neutropenia risks, con-
has received honoraria from Celgene, Janssen-Cilag, and Amgen sequences, and new directions for its management. Cancer 2004; 100:228-37.
23. Miceli T, Colson K, Gavino M, et al. Myelosuppression associated with novel
and has served on the advisory committees of Celgene, Janssen- therapies in patients with multiple myeloma: consensus statement of the IMF Nurse
Cilag, Novartis, and Amgen; Pieter Sonnveld has received re- Leadership Board. Clin J Oncol Nurs 2008; 12:13-20.

10 Clinical Lymphoma, Myeloma & Leukemia February 2012


Antonio Palumbo et al
24. Rajkumar SV, Jacobus S, Callander N, et al. Phase III trial of lenalidomide plus 35. Innes H, Lim SL, Hall A, et al. Management of febrile neutropenia in solid tumours
high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone in and lymphomas using the Multinational Association for Supportive Care in Cancer
newly diagnosed multiple myeloma (E4A03): a trial coordinated by the Eastern (MASCC) risk index: feasibility and safety in routine clinical practice. Support Care
Cooperative Oncology Group. J Clin Oncol 2007; 25(suppl):abstract LBA8025. Cancer 2008; 16:485-91.
Available at: www.asco.org. Accessed November 21, 2011. 36. Cullen M, Baijal S. Prevention of febrile neutropenia: use of prophylactic antibiot-
25. Richardson PG, Sonneveld P, Schuster MW, et al. Bortezomib or high-dose dexa- ics. Br J Cancer 2009;101:S11-4.
methasone for relapsed multiple myeloma. N Engl J Med 2005; 352:2487-98. 37. Sharma A, Lokeshwar N. Febrile neutropenia in haematological malignancies. J
26. Rajkumar SV, Blood E, Vesole D, et al. Phase III clinical trial of thalidomide plus Postgrad Med 2005; 51:S42-8.
dexamethasone compared with dexamethasone alone in newly diagnosed multiple 38. Pagliuca A, Carrington PA, Pettengell R, et al. Guidelines on the use of colony-
myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group. stimulating factors in haematological malignancies. Br J Haematol 2003; 123:22-33.
J Clin Oncol 2006; 24:431-6. 39. Scott SD, Chrischilles EA, Link BK, et al. Days of prophylactic filgrastim use to
27. Palumbo A, Bringhen S, Caravita T, et al. Oral melphalan and prednisone chemo- reduce febrile neutropenia in patients with non-Hodgkin’s lymphoma treated with
therapy plus thalidomide compared with melphalan and prednisone alone in elderly chemotherapy. J Manag Care Pharm 2003; 9:15-21.
patients with multiple myeloma: randomised controlled trial. Lancet 2006; 367: 40. Guidelines for the use of G-CSF following chemotherapy, Surrex, West Sussex and
825-31.
Hampshire cancer network, NHS. Available at: http://www.swshcn.nhs.uk/
28. Oakervee HE, Popat R, Curry N, et al. PAD combination therapy (PS-341/bort-
healthcare-professionals/clinical-policies-and-protocols/supportive-care/g-csf_
ezomib, doxorubicin and dexamethasone) for previously untreated patients with
network_policy. Accessed June 22, 2011.
multiple myeloma. Br J Haematol 2005; 129:755-62.
41. Lieschke GJ, Burgess AW. Granulocyte colony-stimulating factor and granulocyte-
29. San Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and
macrophage colony-stimulating factor. N Engl J Med 1992; 327:28-35.
prednisone for initial treatment of multiple myeloma. N Engl J Med 2008; 359:
906-17. 42. Weycker D, Hackett J, Edelsberg J, et al. Duration of G-CSF therapy and risk of
30. Palumbo A, Gay F. How to treat elderly patients with multiple myeloma: combi- hospitalization for neutropenia or infection. J Clin Oncol 2004; 22(suppl):abstract
nation of therapy or sequencing. Hematology Am Soc Hematol Educ Program 2009: 6731.
566-77. 43. Rolston KV. New trends in patient management: risk-based therapy for febrile
31. Caravita T, Siniscalchi A, Tendas A, et al. Safety and efficacy of a combination patients with neutropenia. Clin Infect Dis 1999; 29:515-21.
therapy with Revlimid, Adriamycin and dexamethasone (RAD) in relapsed/refrac- 44. Adams JR, Angelotta C, Bennett CL. When the risk of febrile neutropenia is 20%,
tory multiple myeloma (MM): a single-centre experience. Ann Hematol 2011; 90: prophylactic colony-stimulating factor use is clinically effective, but is it cost-effec-
115-6. tive? J Clin Oncol 2006; 24:2975-7.
32. Palumbo A, Falco P, Corradini P, et al. Melphalan, prednisone, and lenalidomide 45. Petros WP. Pharmacokinetics and administration of colony-stimulating factors.
treatment for newly diagnosed myeloma: a report from the GIMEMA–Italian Mul- Pharmacotherapy 1992; 12:32S-8S.
tiple Myeloma Network. J Clin Oncol 2007; 25:4459-65. 46. Crawford J, Armitage J, Balducci L, et al. Myeloid growth factors. J Natl Compr
33. Hattori Y, Kakimoto T, Okamoto S, et al. Thalidomide-induced severe neutropenia Canc Network 2009; 7:64-83.
during treatment of multiple myeloma. Int J Hematol 2004; 79:283-8. 47. 2006 update of ASCO Practice Guideline Recommendations for the Use of White
34. Kuderer NM, Dale DC, Crawford J, et al. Mortality, morbidity, and cost associated Blood Cell Growth Factors: Guideline Summary. J Oncol Pract 2006; 2:
with febrile neutropenia in adult cancer patients. Cancer 2006; 106:2258-66. 196-201.

Clinical Lymphoma, Myeloma & Leukemia February 2012 11

You might also like