You are on page 1of 9

Biol Blood Marrow Transplant 19 (2013) 1537e1545

Report

Proceedings from the National Cancer Institutes Second ASBMT


American Society for Blood
and Marrow Transplantation
International Workshop on the Biology, Prevention, and
Treatment of Relapse after Hematopoietic Stem Cell
Transplantation: Part I. Biology of Relapse after
Transplantation
Ronald E. Gress 1, Jeffrey S. Miller 2, Minoo Battiwalla 3, Michael R. Bishop 4,
Sergio A. Giralt 5, Nancy M. Hardy 1, *, Nicolaus Krger 6, Alan S. Wayne 7, 8,
Dan A. Landau 9, Catherine J. Wu 9
From the 1 Experimental Transplantation Immunology Branch, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland
2
Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, Minneapolis, Minnesota
3
Hematology Branch, National Heart, Lung and Blood Institute, Hematology Branch, Bethesda, Maryland
4
Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
5
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
6
Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
7
Pediatric Oncology Branch, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland
8
Current: Childrens Center for Cancer and Blood Diseases, Childrens Hospital Los Angeles, Keck School of Medicine, University of
Southern California, Los Angeles, California
9
Cancer Vaccine Center and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts

Article history: a b s t r a c t
Received 24 August 2013 In the National Cancer Institutes Second Workshop on the Biology, Prevention, and Treatment of Relapse
Accepted 30 August 2013 after Hematopoietic Stem Cell Transplantation, the Scientic/Educational Session on the Biology of Relapse
discussed recent advances in understanding some of the host-, disease-, and transplantation-related
Key Words: contributions to relapse, emphasizing concepts with potential therapeutic implications. Relapse after
Stem cell transplantation hematopoietic stem cell transplantation (HSCT) represents tumor escape, from the cytotoxic effects of the
Relapse conditioning regimen and from immunologic control mediated by reconstituted lymphocyte populations.
Allogeneic
Factors inuencing the biology of the therapeutic graft-versus-malignancy (GVM) effectdand rela-
Prevention
psedinclude conditioning regimen effects on lymphocyte populations and homeostasis, immunologic niches,
Biology
Treatment and the tumor microenvironment; reconstitution of lymphocyte populations and establishment of functional
immune competence; and genetic heterogeneity within the malignancy dening potential for clonal escape.
Recent developments in T cell and natural killer cell homeostasis and reconstitution are reviewed, with
implications for prevention and treatment of relapse, as is the application of modern genome sequencing to
dening the biologic basis of GVM, clonal escape, and relapse after HSCT.
Published by Elsevier Inc. on behalf of American Society for Blood and Marrow Transplantation.

INTRODUCTION processes that depend on recovery of thymic function and


Recovery of immunologic competence contributes to the development of nave and regulatory T cell (Treg) pop-
therapeutic efcacy of hematopoietic stem cell trans- ulations. Peripheral recovery from lymphopenia is marked
plantation (HSCT). T lymphocytes and natural killer (NK) by vulnerability to dysregulated immune responses and
cells have demonstrated antitumor potential in the alloge- immune incompetence. It also represents a period of ther-
neic transplant (AlloSCT) setting and may have therapeutic apeutic opportunity, with potential to direct antigen-driven
benet after autologous transplantation (AHSCT) as well. expansion toward tumor targets.
Successful transplant outcomes require the recovery of
functional immune competence, including reconstitution of
LYMPHOCYTE HOMEOSTASIS: IMPLICATIONS FOR THE
immune effectors from populations transferred in the graft,
PREVENTION AND TREATMENT OF RELAPSE
through peripheral expansion of mature T cells and deriva-
Loss of Lymphocyte Populations with Conditioning
tion of new lymphocyte populations from progenitors, and
A generalized loss of lymphocyte populations is seen with
the establishment of antigen-presenting cell and regulatory
most chemotherapy regimens; such loss can be profound
cell populations. Complete immune reconstitution includes
after even a single cycle of conventional outpatient therapy.
recovery of repertoire and control of autoreactivity,
Myeloablative conditioning regimens are highly lymphoa-
blative, leaving only small residual lymphocyte populations.
Financial disclosure: See Acknowledgments on page 1544. However, after reduced-intensity conditioning, incomplete
* Correspondence and reprint requests: Nancy M. Hardy, MD, Experi- host lymphoid depletion provides for potential expansion of
mental Transplantation and Immunology Branch, National Cancer Institute/
Center for Cancer Research, 10 Center Drive, Hateld Clinical Research
residual host populations along with donor T cell expansion,
Center, Room 3E-3330, Bethesda, MD 20892. adding another layer of complexity to the biology of T cell
E-mail address: hardyn@mail.nih.gov (N.M. Hardy). reconstitution after AlloSCT.
1083-8791/$ e see front matter Published by Elsevier Inc. on behalf of American Society for Blood and Marrow Transplantation.
http://dx.doi.org/10.1016/j.bbmt.2013.08.010
1538 R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545

Limitations to Lymphocyte Recovery after HSCT repertoire diversity, with a high proportion of oligoclonal,
Conditioning expanded CD28 cells by TCR spectratyping that was strongly
Some functional lymphocyte and antigen-presenting cell associated with a history of cytomegalovirus (CMV) infection,
populations are regenerated from graft-derived progenitors suggesting viral antigen-driven peripheral expansion. In
within a few months, including monocytes, dendritic cells, younger adult patients, CD8 T cell recovery bore resem-
and NK cells [1,2]. In the absence of B-lymphocyte directed blance to that of CD4 T cells: phenotypically nave CD8 T
therapy, circulating B cell counts recover within 3 months, cells gradually predominated the circulating CD8 T cell pool
but functional decits persist unless T cell subset recovery during the second year, generating a population with
permits the CD4 T cell help necessary for memory B cell a broadly diverse TCR repertoire and high T cell receptor
development. Memory and effector T cells in the graft, excision circle frequencies resulting from thymic reactivation.
particularly CD8 subsets, can expand in response to anti- A subset of younger patients also demonstrated the early,
genic stimulation [3], but regeneration of T cells with oligoclonal expansion of CD8 T cells, suggesting that
a diverse repertoire of T cell receptors (TCR) requires peripheral and thymic pathways were independent modula-
renewed thymopoiesis. The potential for thymic recovery tors of recovery. A third pattern of CD8 T cell repopulation
after HSCT declines sharply with age [4] and may be was characterized by the absence of both the early expansion
hindered by tissue damagedfrom the conditioning regimen of effectors and the later recovery of nave CD8 T cells; these
and, after AlloSCT, from acute graft-versus-host disease patients experienced prolonged, severe decits in circulating
(GVHD). Resultant thymic epithelial cell damage interferes T cell numbers and in reconstitution of TCR repertoire diver-
with proper clonal deletion of autoreactive T cells and sity in CD4 as well as CD8 T cell subsets (Figure 1). This third
reduces quantitative T cell production, including the gener- pattern exhibits the limits of thymus-independent homeo-
ation of Tregs. Resultant thymic epithelial cell damage may static expansion in reconstituting functional immune
lay the foundation for later development of the competence in middle-aged adults.
autoimmune-like dysfunction of chronic GVHD. The regeneration of Tregs is less well characterized than
for the conventional T cell subsets described. It appears that
General Pathways for Regeneration of T Cell Subsets Treg repopulation also can derive from thymus-dependent
Murine studies identied 2 pathways of T cell regeneration: and peripheral expansion pathways in the setting of lym-
thymic-dependent maturation of new T cells from marrow phopenia [11] and that thymic renewal may yield Treg pop-
progenitors and thymic-independent expansion of mature ulations capable of modulating chronic GVHD [12]. Exogenous
peripheral T cells [5-9]. Through the latter, T cell numbers are IL-2 administration increases the number of circulating Tregs
maintained by a balance between their cytokine consumption through both pathways [13], with therapeutic potential for
and levels of constitutively expressed homeostatic cytokines prevention and treatment of chronic GVHD [14].
(IL-7 and IL-15) that support their maturation, proliferative
expansion, and survival. Homeostatic peripheral expansion in Implications for Prevention and Treatment of Relapse
lymphopenic hosts can be strongly skewed by rapid, antigen- A better understanding of physiologic regulation of
driven proliferation. Translational studies in children, which thymic function is needed to identify clinical strategies to
validated CD45 isoform-dened CD4 T cell subsets, demon- optimize the thymus-dependent pathway, particularly in
strated recovery of total and nave (CD45CD45RO) periph- adults. Insulin-like growth factor 1, androgen blockade,
eral CD4 T cells as early as 6 months after severe and keratinocyte growth factor are candidate regulators
chemotherapy-induced lymphopenia and established thymic- amenable to clinical approaches to enhance recovery of
dependent T cell production as primarily responsible for this thymopoiesis and of functional T cell immunity. Further,
repopulation in young patients [6]. In contrast, in adults, few clinical availability of primary homeostatic cytokines (ie, IL-7
nave peripheral CD4 Tcells are generated during the rst year and IL-15) creates the possibility of enhancing the peripheral
after chemotherapy, whereas circulating CD4 T cells with expansion pathway and tumor antigenedriven expansion
a memory phenotype (CD45RO) recover to nearly pretreat- after HSCT and/or in conjunction with tumor-specic
ment levels (w83%) within 3 months through thymus- immunotherapies. Similarly, our developing understanding
independent peripheral expansion [10]. of the biology of Treg homeostasis may yield opportunity for
Longitudinal study of T cell recovery in adults after mye- relapse prevention through GVHD modulation (eg, IL-2). As
loablative conditioning and AHSCT for breast cancer demon- functionally distinct T cell subsets continue to be identied,
strated the inuence of age-associated thymic renewal on some (eg, memory stem T cells [15]) may be important
CD4 T cell recovery. In patients in whom renewal of thy- determinants of functional immune reconstitution after
mopoiesis was observeddas assessed by nave cell pheno- recovery from lymphopenia. The biology of emerging T cell
type (CD45RA), T cell receptor excision circle frequency, subsets will need to be integrated into our understanding of
thymic size, and rediversication of TCR repertoirednorm- T cell homeostasis and immune reconstitution.
alization of circulating CD4 T cell counts was noted by the Recent advances in cellular engineering create potentially
end of the second year and permitted reestablishment of powerful tools for generating cancer-specic immune
central memory populations. In those who did not reactivate responses. The development of genetically modied effector
thymopoiesis, CD4 T cell counts remained below normal T cells now permits deliberate skewing of the T cell repertoire
even up to 5 years after AHSCT [4]. Recovery of CD8 T cells in toward antitumor effector populations. Chimeric antigen
the same patient cohort followed markedly different repo- receptors redirect T cell specicity and function through
pulation kinetics, with 3 distinct patterns observed (Figure 1). combining a specic antibodys antigen-recognition domain
In more than half of the patients, CD28CD8 T cell expansion with a T cell signaling domain yielding a receptor with high
yielded a massive early spike in total peripheral CD8 T cell specicity and independence from TCReMHC restriction
counts and constituted the dominant population in persis- [16]. The biology of homeostatic antigen-driven peripheral
tently elevated CD8 T cells for 2 years after AHSCT. The expansion may be exploited to favor proliferation of adop-
circulating CD8 T cell population demonstrated limited TCR tively transferred cells; timing administration to coincide
R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545 1539

Figure 1. Diagrammatic representation of the timeline of immune reconstitution in a 40-year-old CMV-seropositive adult. After cytoreductive therapy, the peripheral
CD4 and CD8 T cell populations are severely depleted. The thymus is reduced to a small remnant (shown at 1.5 months post-transplant). CD4 and CD8 T cells
immediately undergo a marked expansion in response to homeostatic cytokines and endogenous antigens, generating a population that is mainly composed of
memory (green) and effector (orange) T cells, with few nave (blue) or T cell receptor excision circle (TREC)ebearing cells (also represented at 1.5 months post-
transplant). The CD4/CD8 ratio becomes inverted by the more rapid expansion of CD8 T cells, demonstrated by the peak of (orange) effector CD8 cells as
compared with the smaller (green) peak of CD4 cells at 1.5 months. TCR repertoire diversity that has been lost by lymphodepletion is further skewed by oligoclonal
expansion of the limited number of remaining cells. The expanded population of CD8 T cells persists and may continue to dominate the CD8 TCR repertoire as shown
in subsequent months by the large effector (orange) CD8 population. Renewed thymopoiesis begins within the rst 6 months, but the full contribution of nave, TREC-
bearing T cells with a diverse TCR repertoire may require 1 to 2 years to be evident. (Reprinted from Williams KM, Hakim FT, Gress RE. T cell immune reconstitution
following lymphodepletion. Semin Immunol 2007;19:318-330, 2007. Copyright (2007), with permission from Elsevier.)

with therapy-induced lymphopenia and/or coadministration AHSCT-associated lymphopenia also provides an ad-
of exogenous homeostatic cytokines are potentially syner- vantageous milieu for homeostatic expansion and an
gistic immunomodulatory strategies to support their survival opportunity to support antigen-directed immunotherapy
and expansion in vivo. Interruption of counter-regulatory by vaccines or other immunomodulatory approaches.
mechanisms may improve antitumor responses as well, Remarkable recent progress in using such strategies was
with Treg depletion and checkpoint inhibition (eg, pro- discussed in Sessions II and III. Perhaps for the rst time in
grammed cell death protein 1 (PD-1) PD-1 [17]) as examples the history of HSCT, we have arrived at an intersection of
of promising strategies. knowledge and technology that may permit removal of the
1540 R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545

fundamental barriers that have limited this eld from its Inhibitory KIR are the main human family of MHC Class I
inception. binding receptors that recognize HLA-Bw4 (present on some
HLA-B and HLA-A alleles) and HLA-C group ligands. KIR are
NK CELL BIOLOGY AND THERAPEUTIC POTENTIAL complex because they can also mediate activation based on
Our understanding of the heterogeneous and complex NK association with adaptor proteins that stimulate function.
cell cytotoxic lymphocyte population evolved from initial Although the KIR family has dominated the clinical literature,
murine transplant studies showing lymphocytes with MHC- the net result of whether or not an NK cell kills its target is
unrestricted cytotoxic antitumor activity in vitro; current determined by a large number of activating and inhibitory
biology incorporates the subsequent identication of interactions independent of MHC binding. Thus, allor-
a distinct NK cell lymphocyte population, NK inhibitory eactivity is determined by a lack of inhibition through Class I
receptors (the LY49 receptor family in mice and inhibitory recognizing receptors and a positive balance of activating
killer immunoglobulin-like receptors [KIR] and NKG2A/CD94 signals including but not limited to activating KIR, CD16
heterodimer in humans), and, more recently, MHC-inde- (FcRgIII), natural cytotoxicity receptors (NKp30, 44, 46),
pendent activating receptors in both mouse and humans. DNAM-1, LFA-1, NKG2C, NKG2D, and CD244 (2B4) (Figure 2).
Ontogeny, homeostatic regulation, and function of NK cells The mechanism by which NK cells acquire self-tolerance
remain areas of active investigation, but it appears they and alloreactivity has been referred to as NK cell education
derive from common lymphoid progenitors in the bone or licensing [21]. Simply stated, this is the process by which
marrow and undergo IL-15edependent development in NK cells acquire function through interactions regulated by
secondary lymphoid organs, with terminal differentiation Class I recognizing inhibitory receptors (inhibitory KIR and
and cytotoxic function acquired in the periphery. Their signaling through NKG2A/CD94 heterodimers that bind HLA-
function is complex, apparently playing important roles in E). In addition to licensing, NK cells can acquire function
both tolerance (ie, MHC-disparate fetal tissue, with an through several activating mechanisms, including cytokine
abundance of CD56-bright, immature NK cells predominat- stimulation and inammation induced by viral infection.
ing the uterine leukocyte population) and surveillance, for Adding to these complexities, specic conditions for acti-
example, killing of tumor and virally infected cells that evade vating components of the KIR family remain largely unde-
T cells via reduced MHC cell-surface expression (with ned; for example, whereas KIR2DS1 recognizes HLA-C2 at
a CD56-dim, terminally differentiated population). Such low afnity [22], the ligand for other activating KIR is
complexity was also rst observed in murine transplant unknown.
models that demonstrated NK cell effects on GVHD modu- Insights from the KIR-homologous murine Ly49 system
lation and graft-versus-malignancy and in inspired human may prove informative. The activating receptor Ly49H
studies that demonstrated NK cell graft-versus-malignancy recognizes the murine CMV glycoprotein m157, providing
activity in acute myelogenous leukemia [18-20]; Dr. Hsu proof-of-principle that activating receptors may recognize
discussed the implications of NK cell immunogenetics for viral proteins [23]. Activation through Ly49H may be effec-
relapse prevention in Session II. tive on NK cells without classic licensing mechanisms.

Figure 2. NK cells express a number of inhibitory and activating receptors that determine function. Some of these interactions are denitively established with
known signaling pathways, whereas others are less clear. Most receptors interact with cellular targets, but CD16 delivers a potent signal by binding the Fc portion of
immunoglobulin-coated targets. The ability of NK cells to kill a target is determined by the net balance of these inhibitory, activating, antibody-dependent, and
adhesion interactions. (Reprinted from Murphy WJ, Parham P, Miller JS. NK cellsdfrom bench to clinic. Biol Blood Marrow Transplant 2012;18:S2-S7. Copyright
(2012), with permission from Elsevier.)
R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545 1541

Ly49H NK cells expand and then contract after viral control, KIR led to exploration of allogeneic NK cell adoptive transfer.
yet these cells can persist in the recipient 3 months after To overcome barriers of allogeneic immunity and achieve
infection, suggesting existence of NK cell memory, further in vivo NK cell expansion, success was dependent on a potent
supported by their heightened responsiveness (such as IFN-g lymphodepleting regimen to create space, increase en-
production) upon secondary challenge [24]. dogenous cytokines, and eliminate immune rejection of
adoptively transferred cells. In 2005, we identied the
Functional Activity of NK Cells Reconstituting Early after requirement for lymphodepleting chemotherapy to promote
Transplantation in vivo expansion of NK cells after adoptive transfer of hap-
After AlloSCT, NK cells are the rst lymphocyte population loidentical related donor NK cells in a nontransplant setting
to recover, with donor cells predominant. NK cells are [29]. NK cells were infused with IL-2 administration for the
attractive to exploit in the setting of HSCT because their early 2-week interval after adoptive transfer. Chimerism studies
recovery occurs when the adaptive immune system is established unequivocally that donor-derived haploidentical
particularly weak. However, the repopulating NK cells that NK cells could persist and expand in vivo in some patients;
are circulating early after AlloSCT are phenotypically and antitumor responses seemed to correlate with in vivo NK cell
functionally different from the highly functional, FcRgIII- expansion.
bearing CD56dim NK cells found in peripheral blood from In some settings, IL-2 administration may result in Treg
healthy donors. These early NK cells may be developmentally expansion and contraction of the immune response [30,31].
immature, with diminished KIR expression and higher In an attempt to further overcome these barriers, some
expression of NKG2A [25] and CD56] CD56bright NK cells are investigations have been exploring increased lymphodeple-
a normally small population in the periphery (usually found tion by adding total body irradiation, approaching myeloa-
in secondary lymphoid tissues and the pregnant uterus); blative intensity conditioning. In this setting, HSCT may be
although they respond to IL-12 and IL-18 with high IFN-g warranted to avoid prolonged neutropenia. Clearly, safer
production, they lack cytotoxic effector capabilities. NK cell strategies are needed to maximize the effector-to-target ratio
responses to targets are defective, especially for target- in vivo. IL-15 administration has recently shown promise in
induced cytokine production [26]. These responses primates; it is being produced by the National Cancer Insti-
normalize over the rst year after HSCT, coinciding with T tute and clinical trials are in progress. The main advantage of
cell recovery and suggesting that T cells may play a role in the IL-15 is that it does not stimulate CD25hi (IL-2Ra) Treg
acquisition of NK cell maturation and cytotoxic function, expansion. An alternative approach is ex vivo expansion of
possibly through the cytokines they release. NK cells, which could permit infusion of a greater cell dose
and, potentially, reduced dependence on potent, toxic
Endogenous Pathways and Pitfalls to Enhanced NK Cell conditioning for lymphodepletion.
Function
Based on murine studies with CMV and Ly49H NK cells, Future Directions in Optimizing NK Cell Therapeutics
human studies suggest that the C-type lectin-like receptor Our understanding of NK celletarget interactions requires
NKG2C and the KIR family may be involved in viral infection. more detailed exploration of the potency of specic NK
We studied NK cell function after human CMV reactivation molecules and their ligands on specic target cells (including
following umbilical cord blood AlloSCT [27]. In this setting, malignant stem cells), the stages of NK maturation, and the
recipient CMV reactivation would recapitulate a primary effect of the recipient milieu on NK cell maturation, survival,
immune response from the CMV-nave donor NK cells. and clonal selection. Strategies to overcome tumor-induced
Patient blood samples were collected at the time of reac- immunosuppression and allogeneic rejection of adoptively
tivation and at 2, 4, and 8 weeks after initiating antiviral transferred cells will likely be needed. Targeting NK cells to
therapy. NKG2C NK cells increased after detection of virus increase their specicity may also enhance efcacy. We have
in the blood, peaking 4 weeks after reactivation. NK cells recently been exploring the potent signal delivered through
expressing NKG2C produced signicantly more IFN-g after CD16] Bi- or trispecic killer cell engagers (BiKEs and
exposure to a Class I- target than NK cells lacking NKG2C. TriKEs)dcomprised of 1 or 2 single-chain Fv for a target-
Four weeks after reactivation, NK cells coexpressing NKG2C specic antigen and a single-chain Fv capable of triggering
and KIR produced signicantly more IFN-g than NK cells CD16dseem to be especially potent for this purpose [32].
expressing KIR but not NKG2C. In the context of education of Further studies will determine the most favorable context for
KIR NK cells through their MHC ligands, only NKG2C NK NK cell therapy, how best to promote in vivo survival needed
cells with self-KIR (ie, NK cells that express KIR where the for clinical response, and how to optimize their specicity in
cognate ligand is expressed in the recipient) made IFN-g; in a minimal residual disease setting after transplantation.
contrast, NKG2C nonself KIR NK cells were hypores-
ponsive. Remarkably, an expanded mature, highly functional INTRATUMORAL HETEROGENEITY AND CLONAL
NKG2CKIR NK cell population persisted throughout the EVOLUTION: DETECTION, IMPACT, AND THERAPEUTIC
rst year after transplant. These ndings support the CHALLENGES
emerging concepts of NK cell memory in humans and that From a clinicians perspective, relapsed hematopoietic
NK cell responses to CMV reactivation may promote more malignant disease is almost uniformly more resistant to
generalized recovery of NK cell maturation and functional therapy than the primary disease. Thus, at the level of the
competence that could be important for protecting against most crucial phenotypic featuredtherapy responsive-
cancer relapse and/or viral infections. nessdrelapsed disease is clearly not the same disease as at
Trials exploring adoptive transfer of autologous activated diagnosis. Given the genetic underpinnings of cancer,
NK cells have not demonstrated efcacy, presumably phenotypic evolution must, at its core, have an underlying
because of inhibition through Class I receptor signaling [28]. genotypic evolution. Originfally theorized by Nowell [33],
Reports of the benet of allogeneic NK cells after AlloSCT genetic instability is expected to lead to heterogeneity as the
with donors expressing nonrecipient (ie, nonself) inhibitory neoplasm progresses, resulting in diverse and genetically
1542 R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545

Figure 3. Potential treatment effects on clonal heterogeneity and disease behavior. Interclonal equilibrium can remain remarkably stable for years (top). However,
when an aggressive minor clone arises (bottom), clonal evolution begins and can be potentially accelerated by therapy. This may be due to differential resistance of
subpopulations to treatment. Additionally, a mass extinction event, such as chemotherapy, may accelerate evolution by removing the strong incumbent and allowing
the tter rising subclone to repopulate the compartment more efciently, as sometimes seen with bottlenecks in population genetics. (Adapted from Landau DA, et al.
Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 2013;152:714-726.)
R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545 1543

distinct subpopulations within the neoplasm. Furthermore, Finally, it should be noted that although not every genetic
a Darwinian selection process can then promote the alteration translates into a phenotypic difference, the accu-
outgrowth of tter subclones, thus constantly reshaping the mulation of large numbers of passenger mutations could
overall tness of the population. Evidence supporting this have more subtle phenotypic implications. Silent mutations
paradigm has accumulated over the past 4 decades with might potentially increase the phenotypic plasticity of cancer
increasing technologic sophistication, including cellular, cells by altering transcriptional networks or epigenetic
cytogenetic, and Sanger sequencing (rst-generation landscapes. They may also act to enhance or suppress the
sequencing) based methods. combined effects of multiple altered genetic elements, which
The advent of next-generation sequencing has now then alter the evolutionary tness of the subclone harboring
enabled the unprecedented ability to systematically discover them.
key genetic alterations that underlie cancer. This technology The understanding that malignant disease is composed of
has been applied across various blood malignancies and has a myriad of diverse subpopulations that have the ability to
revealed the high degree of molecular variation that exists transform and adapt poses a challenge to our traditional
between individuals with the same hematologic malignancy diagnostic schemes. A disease can no longer be dened as
(ie, intertumoral genetic heterogeneity, as reported by Wang a single entity containing a uniform set of genetic abnor-
et al. [34], for example). However, because next-generation malities that is prognostically useful. Of note, reports suggest
sequencing has the potential for sensitive genome-wide that relapse clones could be traced back to minor clones
detection of mutations harbored in only a fraction of the present at initial diagnosis [36,42], suggesting that the
total cell population (as small as .02% [35]), an even more degree of genetic heterogeneity of a tumor is likely to be an
startling discovery has been the detection of pervasive important determinant of therapeutic outcome [43].
genetic heterogeneity within malignant cells of the same This concept of clonal heterogeneity also raises related
individual (ie, intratumoral heterogeneity). and important questions regarding negative consequences of
In analyses using whole genome or whole exome cancer therapies on clonal dynamics and tumor behavior
sequencing, clusters of genetic alterations of similar allelic (Figure 2). In a familiar paradigm, cancer therapy may be
frequency can mark and allow the quantication of distinct actively selective for resistant clones; examples for this are
subpopulations harboring them. Various investigators have numerous, including MSH6 mismatch repair gene mutations
used this strategy to characterize the number and dynamics in recurrent glioblastoma multiforme after treatment with
of subpopulations within diverse blood malignancies temozolomide [41] and the T315I BCR-ABL mutations in
[36,37]. These studies have all demonstrated a high degree chronic myelogenous leukemia [44]. An alternative proc-
of clonal heterogeneity and striking changes in the genetic essd entirely independent of differential tness in the
makeup of the disease on relapse. One important insight context of therapydmay also contribute to the emergence of
arising from these studies is that a branching rather than continuously more aggressive, high-tness subclones. In this
linear pattern of evolution is more commonly observed. setting, massive cell kill after effective cancer therapy, acting
This pattern implies that genetic evolution results from as a classic evolutionary restriction point, would reset
complex tness equilibrium of highly diverse populations interclonal dynamics [45]. Thus, when high-tness sub-
rather than a clear succession of selective sweeps. This clones already exist within a tumor population, treatment
nding is all the more surprising in hematologic malig- could favor subsequent repopulation of the tumor niche with
nancies where the mixing of different cellular compart- a more aggressive subclone, leading to a worse clinical
ments is supposedly higher than in solid malignancies and course and, perhaps, ultimately shorter survival than had no
the mutation rate is lower (Gad Getz, personal communi- treatment been given [46].
cation, Broad Institute of Massachusetts Institute of Tech- Understanding evolutionary dynamics can spark the
nology and Harvard, 2012), both traits that in theory should development of novel therapeutic paradigms. Much atten-
have favored a more linear pattern. tion has been paid to the effectiveness of targeted therapies,
Another insight from these studies is that the coexisting but the consistent observations of clonal evolution provide
subclonal populations also harbor driver lesions that are ex- a cautionary note about their ability to generate enduring
pected to provide tness advantage [38], suggesting that the cancer control. An alternative approach might be to try to
growth of subclones is limited by mutual competition, so- maintain interclonal equilibrium at the expense of maxi-
called clonal interference. Thus, the evolutionary dynamics mizing cell kill [47]. In theory, preservation of sensitive
of tumor subpopulations likely result in the coexistence of clones could, in turn, facilitate continued suppression of
clonal variants. Future studies are anticipated to more fully therapy-resistant clones in a competitive manner. This
address the level of heterogeneity at the microscopic pop- understanding also supports the further renement of
ulation level (eg, by deeper sequencing or by single-cell existing therapies, such as immune-based approaches; in
genomic sequencing) and to what degree those subpopula- essence, this amounts to pitting 1 complex adaptive process
tions interact [39]. Clonal dynamics are illustrated in Figure 3. against another. Here, in the case that immunity is generated
A further layer of complexity to understanding clonal against multiple tumor-specic epitopes (eg, by vaccination
heterogeneity in hematologic malignancies involves the or adoptive cellular therapy), tumor control and eradication
concept of cancer stem cells, in which genetic hierarchy is is achieved through multipronged targeting of subclonal as
inuenced by the hierarchy of tumorigenic capacity. For well as clonal populations. This concept helps us understand
example, in chronic myelogenous leukemia, the sensitivity to why AlloSCT, which relies on mounting donor-derived
imatinib is determined not only by genetic alterations to immunity to eliminate leukemia cells, can be curative even
BCR-ABL but also by decreased sensitivity of leukemic in patients with clonally advanced disease [48].
progenitor cells to this targeted therapy [40]. Therefore,
differentiation states that underlie phenotypic heterogeneity CONCLUSIONS
contribute to the overall clonal diversity and have been It is apparent from these 3 aspects of the biology
linked to crucial clinical outcomes [41]. of relapse after HSCT that with the impressive strides
1544 R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545

made, there are many more questions than answers. A matched-sibling hematopoietic stem cell transplant. Haematologica.
2012;98:346-352.
key objective met at the National Cancer Institutes Second
13. Beyer M, Schumak B, Weihrauch MR, et al. In vivo expansion of naive
Workshop on the Biology, Prevention, and Treatment of CD4 CD25 (high) FOXP3 regulatory T cells in patients with colo-
Relapse after Hematopoietic Stem Cell Transplantation was rectal carcinoma after IL-2 administration. PLoS One. 2012;7:e30422.
the establishment of a consortium of transplant investigators 14. Koreth J, Matsuoka K, Kim HT, et al. Interleukin-2 and regulatory T cells
in graft-versus-host disease. N Engl J Med. 2011;365:2055-2066.
who are keen to work together to further understand the 15. Gattinoni L, Lugli E, Ji Y, et al. A human memory T cell subset with stem
biology of relapse. The Biology Protocol Development Team cell-like properties. Nat Med. 2011;17:1290-1297.
made substantial progress on the development of a multi- 16. Brentjens RJ, Curran KJ. Novel cellular therapies for leukemia: CAR-
modied T cells targeted to the CD19 antigen. Hematology Am Soc
institutional protocol to collect, store, and allocate the crit- Hematol Educ Progr. 2012;2012:143-151.
ical tissue samples necessary to study the basic mechanisms 17. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune
of relapse after AlloSCT. In no other area of post-transplant correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:
2443-2454.
relapse is the need greater for this collective effort than it 18. Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural
is in the biology, in which informative research requires killer cell alloreactivity in mismatched hematopoietic transplants
relapse tissue specimens across multiple transplant plat- [comment]. Science. 2002;295:2097-2100.
19. Cooley S, Weisdorf DJ, Guethlein LA, et al. Donor selection for natural
forms and complex clinical outcomes. Workshop partici- killer cell receptor genes leads to superior survival after unrelated
pants strongly endorsed the Biology Protocol Development transplantation for acute myelogenous leukemia. Blood. 2010;116:
Teams recommendation that the highest priority would be 2411-2419.
20. Venstrom JM, Pittari G, Gooley TA, et al. HLA-C-dependent prevention
to establish a protocol for collection and banking of tumor
of leukemia relapse by donor activating KIR2DS1. N Engl J Med. 2012;
and blood specimens at diagnosis of relapse after AlloSCT, 367:805-816.
along with a procedure for prioritizing research questions 21. Kim S, Poursine-Laurent J, Truscott SM, et al. Licensing of natural killer
and sample allocation. cells by host major histocompatibility complex class I molecules.
Nature. 2005;436:709-713.
22. Winter CC, Gumperz JE, Parham P, et al. Direct binding and functional
transfer of NK cell inhibitory receptors reveal novel patterns of HLA-C
allotype recognition. J Immunol. 1998;161:571-577.
ACKNOWLEDGMENT 23. Arase H, Mocarski ES, Campbell AE, et al. Direct recognition of cyto-
Financial disclosure: Supported in part by the Intramural megalovirus by activating and inhibitory NK cell receptors. Science.
Research Programs of the National Institutes of Health: the 2002;296:1323-1326.
24. Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer
National Cancer Institute/Center for Cancer Research and the cells. Nature. 2009;457:557-561.
National Heart, Lung and Blood Institute. 25. Cooley S, McCullar V, Wangen R, et al. KIR reconstitution is altered by T
Authorship statement: R.E.G., J.S.M., and C.J.W. contributed cells in the graft and correlates with clinical outcomes after unrelated
donor transplantation. Blood. 2005;106:4370-4376.
equally to this work.
26. Foley B, Cooley S, Verneris MR, et al. NK cell education after allogeneic
transplantation: dissociation between recovery of cytokine-producing
and cytotoxic functions. Blood. 2011;118:2784-2792.
REFERENCES 27. Foley B, Cooley S, Verneris MR, et al. Cytomegalovirus reactivation after
1. Boyiadzis M, Memon S, Carson J, et al. Up-regulation of NK cell acti- allogeneic transplantation promotes a lasting increase in educated
vating receptors following allogeneic hematopoietic stem cell trans- NKG2C natural killer cells with potent function. Blood. 2012;119:
plantation under a lymphodepleting reduced intensity regimen is 2665-2674.
associated with elevated IL-15 levels. Biol Blood Marrow Transplant. 28. Burns LJ, Weisdorf DJ, DeFor TE, et al. IL-2-based immunotherapy after
2008;14:290-300. autologous transplantation for lymphoma and breast cancer induces
2. Fagnoni FF, Oliviero B, Giorgiani G, et al. Reconstitution dynamics of immune activation and cytokine release: a phase I/II trial. Bone Marrow
plasmacytoid and myeloid dendritic cell precursors after allogeneic Transplant. 2003;32:177-186.
myeloablative hematopoietic stem cell transplantation. Blood. 2004; 29. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive
104:281-289. transfer and in vivo expansion of human haploidentical NK cells in
3. Peggs KS, Verfuerth S, Pizzey A, et al. Reconstitution of T-cell repertoire patients with cancer. Blood. 2005;105:3051-3057.
after autologous stem cell transplantation: inuence of CD34 selection 30. Geller MA, Cooley S, Judson PL, et al. A phase II study of allogeneic
and cytomegalovirus infection. Biol Blood Marrow Transplant. 2003;9: natural killer cell therapy to treat patients with recurrent ovarian and
198-205. breast cancer. Cytotherapy. 2011;13:98-107.
4. Hakim FT, Memon SA, Cepeda R, et al. Age-dependent incidence, time 31. Bachanova V, Burns LJ, McKenna DH, et al. Allogeneic natural killer cells
course, and consequences of thymic renewal in adults. J Clin Invest. for refractory lymphoma. Cancer Immunol Immunother. 2010;59:
2005;115:930-939. 1739-1744.
5. Mackall CL, Bare CV, Granger LA, et al. Thymic-independent T cell 32. Gleason MK, Verneris MR, Todhunter DA, et al. Bispecic and trispecic
regeneration occurs via antigen-driven expansion of peripheral T cells killer cell engagers directly activate human NK cells through CD16
resulting in a repertoire that is limited in diversity and prone to signaling and induce cytotoxicity and cytokine production. Mol Cancer
skewing. J Immunol. 1996;156:4609-4616. Ther. 2012;11:2674-2684.
6. Mackall CL, Fleisher TA, Brown MR, et al. Age, thymopoiesis, and CD4 33. Nowell PC. The clonal evolution of tumor cell populations. Science.
T-lymphocyte regeneration after intensive chemotherapy. N Engl J Med. 1976;194:23-28.
1995;332:143-149. 34. Wang L, Lawrence MS, Wan Y, et al. SF3B1 and other novel cancer
7. Mackall CL, Fleisher TA, Brown MR, et al. Distinctions between CD8 genes in chronic lymphocytic leukemia. N Engl J Med. 2011;365:
and CD4 T-cell regenerative pathways result in prolonged T-cell 2497-2506.
subset imbalance after intensive chemotherapy. Blood. 1997;89: 35. Campbell P, Pleasance E, Stephens P, et al. Subclonal phylogenetic
3700-3707. structures in cancer revealed by ultra-deep sequencing. Proc Natl Acad
8. Mackall CL, Fleisher TA, Brown MR, et al. Lymphocyte depletion during Sci USA. 2008;105:13081-13086.
treatment with intensive chemotherapy for cancer. Blood. 1994;84: 36. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute
2221-2228. myeloid leukaemia revealed by whole-genome sequencing. Nature.
9. Mackall CL, Gress RE. Pathways of T-cell regeneration in mice and 2012;481:506-510.
humans: implications for bone marrow transplantation and immuno- 37. Egan JB, Shi CX, Tembe W, et al. Whole-genome sequencing of multiple
therapy. Immunol Rev. 1997;157:61-72. myeloma from diagnosis to plasma cell leukemia reveals genomic
10. Hakim FT, Cepeda R, Kaimei S, et al. Constraints on CD4 recovery initiating events, evolution, and clonal tides. Blood. 2012;120:
postchemotherapy in adults: thymic insufciency and apoptotic 1060-1066.
decline of expanded peripheral CD4 cells. Blood. 1997;90:3789-3798. 38. Snuderl M, Fazlollahi L, Le LP, et al. Mosaic amplication of multiple receptor
11. Matsuoka K, Kim HT, McDonough S, et al. Altered regulatory T cell tyrosine kinase genes in glioblastoma. Cancer Cell. 2011;20:810-817.
homeostasis in patients with CD4 lymphopenia following allogeneic 39. Ene CI, Fine HA. Many tumors in one: a daunting therapeutic prospect.
hematopoietic stem cell transplantation. J Clin Invest. 2010;120: Cancer Cell. 2011;20:695-697.
1479-1493. 40. Oravecz-Wilson KI, Philips ST, Yilmaz OH, et al. Persistence of
12. McIver Z, Melenhorst JJ, Wu C, et al. Donor lymphocyte count and leukemia-initiating cells in a conditional knockin model of an imatinib-
thymic activity predict lymphocyte recovery and outcomes after responsive myeloproliferative disorder. Cancer Cell. 2009;16:137-148.
R.E. Gress et al. / Biol Blood Marrow Transplant 19 (2013) 1537e1545 1545

41. Clevers H. The cancer stem cell: premises, promises and challenges. Nat 45. Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple
Med. 2011;17:313-319. myeloma. Nat Rev Cancer. 2012;12:335-348.
42. Mullighan CG, Phillips LA, Su X, et al. Genomic analysis of the clonal origins 46. Landau DA, Stojanov P, Lawrence MS, et al. Shifts in intra-clonal
of relapsed acute lymphoblastic leukemia. Science. 2008;322:1377-1380. dynamics rather than novel mutations are the main engine driving
43. Maley CC, Galipeau PC, Finley JC, et al. Genetic clonal diversity predicts tumor evolution in relapsed CLL. Blood. 2011;118:131-132.
progression to esophageal adenocarcinoma. Nat Genet. 2006;38: 47. Gatenby RA, Silva AS, Gillies RJ, Frieden BR. Adaptive therapy. Cancer
468-473. Res. 2009;69:4894-4903.
44. Roche-Lestienne C, Soenen-Cornu V, Grardel-Duos N, et al. Several 48. Dreger P, Dohner H, Ritgen M, et al. Allogeneic stem cell trans-
types of mutations of the Abl gene can be found in chronic myeloid plantation provides durable disease control in poor-risk chronic
leukemia patients resistant to STI571, and they can pre-exist to the lymphocytic leukemia: long-term clinical and MRD results of the
onset of treatment. Blood. 2002;100:1014-1018. German CLL Study Group CLL3X trial. Blood. 2010;116:2438-2447.

You might also like