Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Nov 1.
Published in final edited form as: Biol Blood Marrow Transplant. 2013 Sep 6;19(11):1537–1545. doi: 10.1016/j.bbmt.2013.08.010

"VSports手机版" Proceedings from the National Cancer Institute’s Second 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 (VSports最新版本) 5, Nancy M Hardy 1, Nicolaus Kröger 6, Alan S Wayne 7,8, Dan A Landau 9, Catherine J Wu (VSports注册入口) 9,*
PMCID: PMC3922045  NIHMSID: NIHMS530902  PMID: 24018395

Abstract

In the National Cancer Institute’s Second Workshop on the Biology, Prevention, and Treatment of Relapse After Hematopoietic Stem Cell Transplantation, the Scientific/Educational Session on the Biology of Relapse discussed recent advances in understanding some of the host, disease and transplant-related contributions to relapse, emphasizing concepts with potential therapeutic implications. Relapse after hematopoietic stem cell transplantation (HSCT) represents tumor escape – from the cytotoxic effects of the conditioning regimen and from immunologic control mediated by reconstituted lymphocyte populations. Factors influencing the biology of the therapeutic graft-versus-malignancy (GVM) effect – and relapse – include conditioning regimen effects upon lymphocyte populations and homeostasis, immunologic niches, and the tumor microenvironment; reconstitution of lymphocyte populations and establishment of functional immune competence; and genetic heterogeneity within the malignancy defining potential for clonal escape VSports最新版本. Recent developments in T- and NK-cell homeostasis and reconstitution are reviewed, with implications for prevention and treatment of relapse, as is the application of modern genome sequencing to defining the biologic basis of GVM, clonal escape and relapse after HSCT.

INTRODUCTION

Recovery of immunologic competence contributes to the therapeutic efficacy of hematopoietic stem cell transplantation (HSCT) V体育平台登录. T-lymphocytes and natural killer (NK) cells have demonstrated antitumor potential in the allogeneic transplant (AlloSCT) setting and may have therapeutic benefit after autologous transplantation (AHSCT) as well. Successful transplant outcomes require the recovery of functional immune competence, including reconstitution of immune effectors from populations transferred in the graft, through peripheral expansion of mature T cells and derivation of new lymphocyte populations from progenitors, as well as the establishment of antigen-presenting cell (APC) and regulatory-cell populations. Complete immune reconstitution includes recovery of repertoire and control of autoreactivity, processes that depend upon recovery of thymic function and development of naïve and regulatory T cell (Treg) populations. Peripheral recovery from lymphopenia is marked by vulnerability to dysregulated immune responses and immune incompetence. It also represents a period of therapeutic opportunity, with potential to direct antigen-driven expansion toward tumor targets.

I. LYMPHOCYTE HOMEOSTASIS: IMPLICATIONS FOR THE PREVENTION AND TREATMENT OF RELAPSE

Loss of Lymphocyte Populations With Conditioning

There is a generalized loss of lymphocyte populations with most chemotherapy regimens; such loss can be profound after even a single cycle of conventional outpatient therapy. Myeloablative conditioning regimens are highly lymphoablative, leaving only small residual lymphocyte populations. However, following reduced-intensity conditioning, incomplete host lymphoid depletion provides for potential expansion of residual host populations along with donor T cell expansion, adding another layer of complexity to the biology of T cell reconstitution following AlloSCT VSports注册入口.

Limitations to Lymphocyte Recovery After HSCT Conditioning

Some functional lymphocyte and APC populations are regenerated from graft-derived progenitors within a few months, including monocytes, dendritic cells, and NK cells (1, 2). In the absence of B-lymphocyte directed therapy, circulating B cell counts recover within three months, but functional deficits will persist unless T cell subset recovery permits the CD4+ T cell help necessary for memory B cell development. Memory and effector T cells in the graft, particularly CD8+ subsets, can expand in response to antigenic stimulation (3), but regeneration of T cells with a diverse repertoire of T cell receptors (TCR) requires renewed thymopoiesis V体育官网入口. The potential for thymic recovery after HSCT declines sharply with age (4) and may be hindered by tissue damage - from the conditioning regimen as well as, after AlloSCT, acute graft-versus-host disease (GVHD). Resultant thymic epithelial cell (TEC) damage interferes with proper clonal deletion of autoreactive T cells and reduces quantitative T cell production, including the generation of Tregs. Resultant TEC damage may lay the foundation for later development of the autoimmune-like dysfunction of chronic GVHD.

General Pathways for Regeneration of T Cell Subsets (V体育安卓版)

Murine studies identified two pathways of T cell regeneration: thymic-dependent maturation of new T cells from marrow progenitors and thymic-independent expansion of mature peripheral T cells (5–9). Through the latter, T cell numbers are maintained by a balance between their cytokine consumption and levels of constitutively expressed homeostatic cytokines (IL-7 and IL-15) that support their maturation, proliferative expansion and survival. Homeostatic peripheral expansion in lymphopenic hosts can be strongly skewed by rapid, antigen-driven proliferation VSports在线直播. Translational studies in children, which validated CD45 isoform-defined CD4+ T cell subsets, demonstrated recovery of total and naïve (CD45+CD45RO−) peripheral CD4+ T cells as early as six months following severe chemotherapy-induced lymphopenia, and established thymic-dependent T cell production as primarily responsible for this repopulation in young patients (6). In contrast, in adults, few naïve peripheral CD4+ T cells are generated during the first year after chemotherapy, while circulating CD4+ T cells with a memory phenotype (CD45RO+) recover to nearly pretreatment levels (~83%) within three months through thymus-independent peripheral expansion (10).

Longitudinal study of T cell recovery in adults after myeloablative conditioning and AHSCT for breast cancer demonstrated the influence of age-associated thymic renewal on CD4+ T cell recovery. In patients in whom renewal of thymopoiesis was observed – as assessed by naïve cell phenotype (CD45RA+), T cell receptor excision circle (TREC) frequency, thymic size and rediversification of TCR repertoire – normalization of circulating CD4+ T cell counts was noted by the end of the second year and permitted reestablishment of central memory populations. In those who did not reactivate thymopoiesis, CD4+ T cell counts remained below normal even up to five years after AHSCT (4). Recovery of CD8+ T cells in the same patient cohort followed markedly different repopulation kinetics, with three distinct patterns observed (Figure 1). In more than half of the patients, CD28−CD8+ T cell expansion yielded a massive early spike in total peripheral CD8+ T cell counts, and constituted the dominant population in persistently elevated CD8+ T cells for two years after AHSCT. The circulating CD8+ T cell population demonstrated limited TCR repertoire diversity, with a high proportion of oligoclonal, expanded, CD28− cells by TCR spectratyping that was strongly associated with a history of CMV infection, suggesting viral antigen-driven peripheral expansion. In younger adult patients, CD8+ T cell recovery bore resemblance to that of CD4+ T cells: phenotypically naïve CD8+ T cells gradually predominated the circulating CD8+ T cell pool during the second year, generating a population with a broadly diverse TCR repertoire and high TREC frequencies resulting from thymic reactivation. A subset of younger patients also demonstrated the early, oligoclonal expansion of CD8+ T cells, suggesting that peripheral and thymic pathways were independent modulators of recovery. A third pattern of CD8+ T cell repopulation was characterized by the absence of both the early expansion of effectors and the later recovery of naïve CD8+ T cells; these patients experienced prolonged, severe deficits in circulating T cell numbers and in reconstitution of TCR repertoire diversity in CD4+ as well as CD8+ T-cell subsets (Fig. 1). This third pattern exhibits the limits of thymus-independent homeostatic expansion in reconstituting functional immune competence in middle-aged adults V体育2025版.

Figure 1. Diagrammatic representation of the timeline of immune reconstitution in a 40 year old, CMV seropositive adult.

Figure 1

Following 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 month 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 naïve (blue) or TREC-bearing cells (also represented at 1. 5 month 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 to the smaller (green) peak of CD4+ cells at 1 VSports. 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 first 6 months, but the full contribution of naïve, TREC-bearing T cells with a diverse TCR repertoire may require 1 – 2 years to be evident. *Reprinted from Seminars in Immunology, Vol 19 No 5, K. M. Williams, F. T. Hakim, R. E. Gress, T cell immune reconstitution following lymphodepletion, 318-30, 2007, Copyright (2007), with permission from Elsevier.

The regeneration of Tregs is less well characterized than for the conventional T cell subsets described. It appears that Treg repopulation also can derive from thymus-dependent and peripheral expansion pathways in the setting of lymphopenia (11), and that thymic renewal may yield Treg populations capable of modulating chronic GVHD (12). Exogenous IL-2 administration increases the number of circulating Tregs through both pathways (13), with therapeutic potential for prevention and treatment of chronic GVHD (14).

Implications for the Prevention and Treatment of Relapse

A better understanding of physiologic regulation of thymic function is needed to identify clinical strategies to optimize the thymus-dependent pathway, particularly in adults. Insulin-like growth factor-1, androgen blockade and keratinocyte growth factor are candidate regulators amenable to clinical approaches to enhance recovery of thymopoiesis and of functional T cell immunity. Further, clinical availability of primary homeostatic cytokines (i.e., IL-7 and IL-15) (21) creates the possibility of enhancing the peripheral expansion pathway and tumor-antigen driven expansion after HSCT and/or in conjunction with tumor-specific immunotherapies. Similarly, our developing understanding of the biology of Treg homeostasis may yield opportunity for relapse prevention through GVHD modulation (e.g., IL-2). As functionally distinct T cell subsets continue to be identified, some, e.g., “memory stem T cells” (15), may be important determinants of functional immune reconstitution following recovery from lymphopenia. The biology of emerging T cell subsets will need to be integrated into our understanding of T cell homeostasis and immune reconstitution.

Recent advances in cellular engineering create potentially powerful tools for generating cancer-specific immune responses. The development of genetically modified effector T cells now permits deliberate skewing of the T cell repertoire toward antitumor effector populations. Chimeric antigen receptors redirect T cell specificity and function through combining a specific antibody’s antigen-recognition domain with a T cell signaling domain yielding a receptor with high specificity and independence from TCR-MHC restriction (16). The biology of homeostatic antigen-driven peripheral expansion may be exploited to favor proliferation of adoptively transferred cells; timing administration to coincide with therapy-induced lymphopenia and/or coadministration of exogenous homeostatic cytokines are potentially synergistic immunomodulatory strategies to support their survival and expansion in vivo. Interruption of counter-regulatory mechanisms may improve antitumor responses as well, with Treg depletion and checkpoint inhibition (e.g., PD-1 (17)) being examples of promising strategies. AHSCT-associated lymphopenia also provides an advantageous milieu for homeostatic expansion and an opportunity to support antigen-directed immunotherapy by vaccines or other immunomodulatory approaches. Remarkable recent progress in employing such strategies was discussed in Sessions II and III (4950). Perhaps for the first time in the history of hematopoietic stem cell transplantation, we have arrived at an intersection of knowledge and technology that may permit removal of the fundamental barriers that have limited this field from its inception.

II. NATURAL KILLER CELL BIOLOGY AND THERAPEUTIC POTENTIAL

Our understanding of the heterogeneous and complex natural killer (NK) cell cytotoxic lymphocyte population evolved from initial murine transplant studies showing lymphocytes with MHC-unrestricted cytotoxic antitumor activity in vitro; current biology incorporates the subsequent identification of a distinct NK cell lymphocyte population, NK inhibitory receptors (the LY49 receptor family in mice and inhibitory killer immunoglobulin-like receptors (KIR) and NKG2A/CD94 heterodimer in humans) and, more recently, MHC-independent activating receptors in both mouse and man. Ontogeny, homeostatic regulation and function of NK cells remain areas of active investigation, but it appears that they derive from common lymphoid progenitors in the bone marrow, undergo IL-15 dependent development in secondary lymphoid organs, with terminal differentiation and cytotoxic function acquired in the periphery. Their function is complex, apparently playing important roles in both tolerance, i.e., MHC-disparate fetal tissue (with an abundance of CD56-bright, immature NK cells predominating the uterine leukocyte population) and surveillance, e.g., killing of tumor and virally infected cells that evade T cells via reduced MHC cell-surface expression (with a CD56-dim, terminally differentiated population). Such complexity was also first observed in murine transplant models that demonstrated NK cell effects on GVHD modulation and GVM, and inspired human studies that demonstrated NK cell GVM activity in acute myelogenous leukemia (AML) (1820); Dr. Hsu discussed the implications of NK-cell immunogenetics for relapse prevention in Session II (49).

Inhibitory KIR are the main human family of MHC Class I binding receptors that recognize HLA-Bw4 (present on some HLA-B and HLA-A alleles) and HLA-C group ligands. KIR are complex because they can also mediate activation based on association with adaptor proteins that stimulate function. While the KIR family has dominated the clinical literature, the net result of whether or not an NK cell kills its target is determined by a large number of activating and inhibitory interactions independent of MHC binding. Thus, alloreactivity is determined by a lack of inhibition through Class I recognizing receptors and a positive balance of activating signals including but not limited to: activating KIR, CD16 (FcRγIII), natural cytotoxicity receptors (NKp30, 44, 46), DNAM-1, LFA-1, NKG2C, NKG2D, and CD244 (2B4) (Figure 2).

Figure 2. NK cells express a number of inhibitory and activating receptors that determine function.

Figure 2

Some of these interactions are definitively established with known signaling pathways, while 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 Biology of Blood and Marrow Transplantation, Vol 18 (1), S2–7, W.J. Murphy, P. Parham, J.S. Miller, NK cells – from bench to clinic, S2–7, 2012, Copyright (2012), with permission from Elsevier.

The mechanism by which NK cells acquire self-tolerance and alloreactivity has been referred to as NK cell education or licensing (21). Simply stated, this is the process by which NK cells acquire function through interactions regulated by Class I recognizing inhibitory receptors (inhibitory KIR and signaling through NKG2A/CD94 heterodimers that bind HLA-E). In addition to licensing, NK cells can acquire function through several activating mechanisms including cytokine stimulation and inflammation induced by viral infection. Adding to these complexities, specific conditions for activating components of the KIR family remain largely undefined; for example, while KIR2DS1 recognizes HLA-C2 at low affinity (22), the ligand for other activating KIR is unknown.

Insights from the KIR-homologous murine Ly49 system may prove informative. The activating receptor Ly49H recognizes the murine cytomegalovirus (CMV) glycoprotein m157, providing “proof-of-principle” that activating receptors may recognize viral proteins (23). Activation through Ly49H may be effective on NK cells without classic licensing mechanisms. Ly49H+ NK cells expand and then contract after viral control, yet these cells can persist in the recipient three months after infection, suggesting existence of “NK cell memory,” further supported by their heightened responsiveness (such as interferon-γ (IFNγ) production) upon secondary challenge (24).

"V体育2025版" Functional Activity of NK Cells Reconstituting Early After Transplantation

Following AlloSCT, NK cells are the first lymphocyte population to recover, with donor cells predominant. NK cells are attractive to exploit in the setting of HSCT because their early recovery occurs when the adaptive immune system is particularly weak. However, the repopulating NK cells that are circulating early after AlloSCT are phenotypically and functionally different from the highly functional, FcRγIII-bearing CD56dim NK cells found in peripheral blood from healthy donors. These early NK cells may be developmentally immature, with diminished KIR expression and higher expression of NKG2A (25) and CD56. CD56bright NK cells are a normally small population in the periphery (usually found in secondary lymphoid tissues and the pregnant uterus); while they respond to IL-12 and IL-18 with high IFNγ production, they lack cytotoxic effector capabilities. NK cell responses to targets are defective, especially for target-induced cytokine production (26). These responses normalize over the first year following HSCT, coinciding with T cell recovery and suggesting that T cells may play a role in the acquisition of NK cell maturation and cytotoxic function, possibly through the cytokines they release.

Endogenous Pathways and Pitfalls to Enhanced NK Cell Function

Based on murine studies with CMV and Ly49H+ NK cells, human studies suggest that the C-type lectin-like receptor NKG2C and the KIR family may be involved in viral infection. We have studied NK cell function after human CMV reactivation following umbilical cord blood (UCB) AlloSCT (27). In this setting, recipient CMV reactivation would recapitulate a primary immune response from the CMV-naïve donor NK cells. Patient blood samples were collected at the time of reactivation and at two, four and eight weeks after initiating antiviral therapy. NKG2C+ NK cells increased following detection of virus in the blood, peaking four weeks after reactivation. NK cells expressing NKG2C produced significantly more IFNγ after exposure to a Class I target than NK cells lacking NKG2C. Four weeks after reactivation, NK cells coexpressing NKG2C and KIR produced significantly more IFNγ than NK cells expressing KIR but not NKG2C. In the context of education of KIR+ NK cells through their MHC ligands, only NKG2C+ NK cells with self-KIR (i.e., NK cells that express KIR where the cognate ligand is expressed in the recipient) made IFNγ; in contrast, NKG2C+ nonself KIR+ NK cells were hyporesponsive. Remarkably, an expanded mature, highly functional NKG2C+KIR+ NK cell population persisted throughout the first year after transplant. These findings support the emerging concepts of NK cell memory in humans and that NK cell responses to CMV reactivation may promote more generalized recovery of NK cell maturation and functional competence that could be important for protecting against cancer relapse and/or viral infections.

Trials exploring adoptive transfer of autologous activated NK cells have not demonstrated efficacy, presumably because of inhibition through Class I receptor signaling (28). Reports of the benefit of allogeneic NK cells after AlloSCT with donors expressing nonrecipient (i.e., nonself) inhibitory KIR led to exploration of allogeneic NK cell adoptive transfer. In order to overcome barriers of allogeneic immunity and achieve in-vivo NK cell expansion, success was dependent on a potent lymphodepleting regimen to 1) create space; 2) increase endogenous cytokines; and 3) to eliminate immune rejection of adoptively transferred cells. In 2005, we identified the requirement for lymphodepleting chemotherapy to promote in-vivo expansion of NK cells after adoptive transfer of haploidentical related-donor NK cells in a nontransplant setting (29). NK cells were infused with IL-2 administration for the two-week interval after adoptive transfer. Chimerism studies established unequivocally that donor-derived haploidentical NK cells could persist and expand in vivo in some patients; antitumor responses seemed to correlate with in-vivo NK cell expansion. In some settings, IL-2 administration may result in Treg expansion and contraction of the immune response (30, 31). In an attempt to further overcome these barriers, some investigations have been exploring increased lymphodepletion by adding total body irradiation, approaching myeloablative intensity conditioning. In this setting, HSCT may be warranted to avoid prolonged neutropenia. Clearly, safer strategies are needed to maximize the effector-to-target ratio in vivo. IL-15 administration has recently shown promise in primates; it is being produced by the NCI and clinical trials are in progress. The main advantage of IL-15 is that it does not stimulate CD25hi (IL-2Rα) Treg expansion. An alternative approach is ex-vivo expansion of NK cells, which could permit infusion of a greater cell dose and, potentially, reduced dependence upon potent, toxic conditioning for lymphodepletion.

Future Directions in Optimizing NK Cell Therapeutics

Our understanding of NK cell-target interactions requires more detailed exploration of the potency of: specific NK molecules and their ligands on specific target cells (including malignant stem cells); the stages of NK maturation; and the effect of the recipient milieu on NK cell maturation, survival and clonal selection. Strategies to overcome tumor-induced immunosuppression and allogeneic rejection of adoptively transferred cells will likely be needed. Targeting NK cells to increase their specificity may also enhance efficacy. We have recently been exploring the potent signal delivered through CD16. Bi- or tri-specific killer-cell engagers (“BiKEs” and “TriKEs”) - comprised of one or two single-chain Fv for a target-specific antigen and a single-chain Fv capable of triggering CD16 - seem to be especially potent for this purpose (32). Further studies will determine the most favorable context for NK cell therapy, how best to promote in-vivo survival needed for clinical response and how to optimize their specificity in a minimal-residual-disease setting after transplantation.

III. INTRATUMORAL HETEROGENEITY AND CLONAL EVOLUTION: DETECTION, IMPACT AND THERAPEUTIC CHALLENGES

From a clinician’s perspective, relapsed hematopoietic malignant disease is almost uniformly more resistant to therapy than the primary disease. Thus, at the level of the most crucial phenotypic feature—therapy responsiveness—relapsed disease is clearly not the same disease as at diagnosis. Given the genetic underpinnings of cancer, phenotypic evolution must, at its core, have an underlying genotypic evolution. Originally theorized by Nowell, genetic instability is expected to lead to heterogeneity as the neoplasm progresses (33), resulting in diverse and genetically distinct subpopulations within the neoplasm. Furthermore, a Darwinian selection process can then promote the outgrowth of fitter subclones, thus constantly reshaping the overall fitness of the population. Evidence supporting this paradigm has accumulated over the past four decades with increasing technological sophistication, including cellular, cytogenetic and Sanger sequencing (“first-generation sequencing”) based methods.

The advent of next-generation sequencing (NGS) has now enabled the unprecedented ability to systematically discover key genetic alterations that underlie cancer. This technology has been applied across various blood malignancies and has revealed the high degree of molecular variation that exists between individuals with the same hematologic malignancy (i.e., intertumoral genetic heterogeneity, as reported by Wang and colleagues, for example (34)). However, because NGS has the potential for sensitive genome-wide detection of mutations harbored in only a fraction of the total cell population (as small as 0.02% (35)), an even more startling discovery has been the detection of pervasive genetic heterogeneity within malignant cells of the same individual (i.e., intratumoral heterogeneity).

In analyses using whole-genome or -exome sequencing, clusters of genetic alterations of similar allelic frequency can mark and allow the quantification of distinct subpopulations harboring them. Various investigators have used this strategy to characterize the number and dynamics of subpopulations within diverse blood malignancies (36, 37). These studies have all demonstrated a high degree of clonal heterogeneity and striking changes in the genetic makeup of the disease upon relapse. One important insight arising from these studies is that a branching rather than linear pattern of evolution is more commonly observed. This pattern implies that genetic evolution results from complex fitness equilibrium of highly diverse populations rather than a clear succession of selective sweeps. This finding is all the more surprising in hematologic malignancies where the mixing of different cellular compartments is supposedly higher than in solid malignancies and the mutation rate is lower (Gad Getz, personal communication) – both traits that in theory should have favored a more linear pattern. Another insight from these studies is that the coexisting subclonal populations also harbor driver lesions that are expected to provide fitness advantage (38), suggesting that the growth of subclones is limited by mutual competition – so-called clonal interference. Thus the evolutionary dynamics of tumor subpopulations likely result in the coexistence of clonal variants. Future studies are anticipated to more fully address the level of heterogeneity at the microscopic population level (e.g., by deeper sequencing or by single-cell genomic sequencing) and to what degree those subpopulations interact (39). Clonal dynamics are illustrated in Figure 3.

Figure 3. Potential treatment effects on clonal heterogeneity and disease behavior.

Figure 3

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 fitter rising subclone to repopulate the compartment more efficiently, as sometimes seen with bottlenecks in population genetics. Adapted from Landau, D.A., et al., Evolution and impact of subclonal mutations in chronic lymphocytic leukemia, Cell, Vol 152 (4), 714–26, 2013.

A further layer of complexity to understanding clonal heterogeneity in hematologic malignancies involves the concept of cancer stem cells, in which genetic hierarchy is influenced by the hierarchy of tumorigenic capacity. For example, in chronic myelogenous leukemia, the sensitivity to imatinib is determined not only by genetic alterations to BCR-ABL, but also by decreased sensitivity of leukemic progenitor cells to this targeted therapy (40). Therefore, differentiation states that underlie phenotypic heterogeneity contribute to the overall clonal diversity and have been linked to crucial clinical outcomes (41).

Finally, it should be noted that although not every genetic alteration will translate into a phenotypic difference, the accumulation of large numbers of passenger mutations could have more subtle phenotypic implications. Silent mutations might potentially increase the phenotypic plasticity of cancer cells by altering transcriptional networks or epigenetic landscapes. They may also act to enhance or suppress the combined effects of multiple altered genetic elements, which then alter the evolutionary fitness of the subclone harboring them.

The understanding that malignant disease is composed of a myriad of diverse subpopulations that have the ability to transform and adapt poses a challenge to our traditional diagnostic schemes. A disease can no longer be defined as a single entity containing a uniform set of genetic abnormalities that is prognostically useful. Of note, recent reports suggest that relapse clones could be traced back to minor clones present at initial diagnosis (36, 42), suggesting that the degree of genetic heterogeneity of a tumor is likely to be an important determinant of therapeutic outcome (43).

This concept of clonal heterogeneity also raises related and important questions regarding negative consequences of cancer therapies on clonal dynamics and tumor behavior (Figure 3). In a familiar paradigm, cancer therapy may be actively selective for resistant clones; examples for this are numerous, including MSH6 mismatch repair gene mutations in recurrent glioblastoma multiforme after treatment with temozolomide (41) and the T315I BCR-ABL mutations in chronic myelogenous leukemia (44). An alternative process - entirely independent of differential fitness in the context of therapy - may also contribute to the emergence of continuously more aggressive, high-fitness subclones. In this setting, massive cell kill following effective cancer therapy, acting as a classic evolutionary restriction point, would reset interclonal dynamics (45). Thus, when high-fitness subclones already exist within a tumor population, treatment could favor subsequent repopulation of the tumor niche with a more aggressive subclone, leading to a worse clinical course and, perhaps, ultimately shorter survival than had no treatment been given (46).

Understanding evolutionary dynamics can spark the development of novel therapeutic paradigms. Much attention has been paid to the effectiveness of targeted therapies, but the consistent observations of clonal evolution provide a cautionary note about their ability to generate enduring cancer control. An alternative approach might be to try to maintain interclonal equilibrium at the expense of maximizing cell kill (47). In theory, preservation of sensitive clones could, in turn, facilitate continued suppression of therapy-resistant clones in a competitive manner. This understanding also supports the further refinement of existing therapies, such as immune-based approaches; in essence, this amounts to pitting one complex adaptive process against another. Here, in the case that immunity is generated against multiple tumor-specific epitopes (e.g., by vaccination or adoptive cellular therapy), tumor control and eradication is achieved through multipronged targeting of subclonal as well as clonal populations. This concept helps us understand why AlloSCT, which relies on mounting donor-derived immunity to eliminate leukemia cells, can be curative even in patients with clonally advanced disease (48).

CONCLUSIONS

It is apparent from these three aspects of the biology of relapse after HSCT that, while there have been impressive strides, there are many more questions than answers. A key objective met at the National Cancer Institute’s Second Workshop on the Biology, Prevention, and Treatment of Relapse After Hematopoietic Stem Cell Transplantation was the establishment of a consortium of transplant investigators who are keen to work together to further understand the biology of relapse. The Biology Protocol Development Team made substantial progress on the development of a multi-institutional protocol to collect, store and allocate the critical tissue samples necessary to study the basic mechanisms of relapse after AlloSCT. In no other area of posttransplant relapse is the need greater for this collective effort than it is in the biology, in which informative research requires relapse tissue specimens across multiple transplant platforms and complex clinical outcomes. Workshop participants strongly endorsed the Biology Protocol Development Team’s recommendation that the highest priority be to establish a protocol for collection and banking of tumor and blood specimens at diagnosis of relapse after AlloSCT, along with a procedure for prioritizing research questions and sample allocation.

Acknowledgments (V体育ios版)

This work was supported in part by the Intramural Research Programs of the National Institute of Health: the National Cancer Institute/Center for Cancer Research and the National Heart, Lung and Blood Institute.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Boyiadzis M, Memon S, Carson J, et al. Up-regulation of NK cell activating receptors following allogeneic hematopoietic stem cell transplantation under a lymphodepleting reduced intensity regimen is associated with elevated IL-15 levels. Biol Blood Marrow Transplant. 2008;14:290–300. doi: 10.1016/j.bbmt.2007.12.490. [DOI] [PubMed] [Google Scholar]
  • 2.Fagnoni FF, Oliviero B, Giorgiani G, et al. Reconstitution dynamics of plasmacytoid and myeloid dendritic cell precursors after allogeneic myeloablative hematopoietic stem cell transplantation. Blood. 2004;104:281–289. doi: 10.1182/blood-2003-07-2443. [DOI] [PubMed] [Google Scholar]
  • 3.Peggs KS, Verfuerth S, Pizzey A, et al. Reconstitution of T-cell repertoire after autologous stem cell transplantation: influence of CD34 selection and cytomegalovirus infection. Biol Blood Marrow Transplant. 2003;9:198–205. doi: 10.1053/bbmt.2003.50010. ["V体育安卓版" DOI] [PubMed] [Google Scholar]
  • 4.Hakim FT, Memon SA, Cepeda R, et al. Age-dependent incidence, time course, and consequences of thymic renewal in adults. J Clin Invest. 2005;115:930–939. doi: 10.1172/JCI22492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mackall CL, Bare CV, Granger LA, Sharrow SO, Titus JA, Gress RE. Thymic-independent T cell regeneration occurs via antigen-driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing. J Immunol. 1996;156:4609–4616. ["V体育官网入口" PubMed] [Google Scholar]
  • 6.Mackall CL, Fleisher TA, Brown MR, et al. Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy. N Engl J Med. 1995;332:143–149. doi: 10.1056/NEJM199501193320303. [DOI] [PubMed] [Google Scholar]
  • 7.Mackall CL, Fleisher TA, Brown MR, et al. Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy. Blood. 1997;89:3700–3707. ["V体育安卓版" PubMed] [Google Scholar]
  • 8.Mackall CL, Fleisher TA, Brown MR, et al. Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood. 1994;84:2221–2228. ["V体育安卓版" PubMed] [Google Scholar]
  • 9.Mackall CL, Gress RE. Pathways of T-cell regeneration in mice and humans: implications for bone marrow transplantation and immunotherapy. Immunol Rev. 1997;157:61–72. doi: 10.1111/j.1600-065x.1997.tb00974.x. ["VSports手机版" DOI] [PubMed] [Google Scholar]
  • 10.Hakim FT, Cepeda R, Kaimei S, et al. Constraints on CD4 recovery postchemotherapy in adults: thymic insufficiency and apoptotic decline of expanded peripheral CD4 cells. Blood. 1997;90:3789–3798. ["VSports手机版" PubMed] [Google Scholar]
  • 11.Matsuoka K, Kim HT, McDonough S, et al. Altered regulatory T cell homeostasis in patients with CD4+ lymphopenia following allogeneic hematopoietic stem cell transplantation. J Clin Invest. 2010;120:1479–1493. doi: 10.1172/JCI41072. [VSports手机版 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.McIver Z, Melenhorst JJ, Wu C, et al. Donor lymphocyte count and thymic activity predict lymphocyte recovery andoutcomes after matched-sibling hematopoietic stem cell transplant. Haematologica. 2012 doi: 10.3324/haematol.2012.072991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Beyer M, Schumak B, Weihrauch MR, et al. In vivo expansion of naive CD4+ CD25(high) FOXP3+ regulatory T cells in patients with colorectal carcinoma after IL-2 administration. PLoS One. 2012;7:e30422. doi: 10.1371/journal.pone.0030422. [DOI (VSports)] [PMC free article] [PubMed] [Google Scholar]
  • 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. doi: 10.1056/NEJMoa1108188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gattinoni L, Lugli E, Ji Y, et al. A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17:1290–1297. doi: 10.1038/nm.2446. [DOI (V体育2025版)] [PMC free article] [PubMed] [Google Scholar]
  • 16.Brentjens RJ, Curran KJ. Novel cellular therapies for leukemia: CAR-modified T cells targeted to the CD19 antigen. Hematology Am Soc Hematol Educ Program. 2012;2012:143–151. doi: 10.1182/asheducation-2012.1.143. [DOI (VSports最新版本)] [PMC free article] [PubMed] [Google Scholar]
  • 17.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants.[comment] Science. 2002;295:2097–2100. doi: 10.1126/science.1068440. [DOI (VSports注册入口)] [PubMed] [Google Scholar]
  • 19.Cooley S, Weisdorf DJ, Guethlein LA, et al. Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia. Blood. 2010;116:2411–2419. doi: 10.1182/blood-2010-05-283051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Venstrom JM, Pittari G, Gooley TA, et al. HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1. N Engl J Med. 2012;367:805–816. doi: 10.1056/NEJMoa1200503. ["V体育平台登录" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kim S, Poursine-Laurent J, Truscott SM, et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature. 2005;436:709–713. doi: 10.1038/nature03847. [DOI (VSports最新版本)] [PubMed] [Google Scholar]
  • 22.Winter CC, Gumperz JE, Parham P, Long EO, Wagtmann N. Direct binding and functional transfer of NK cell inhibitory receptors reveal novel patterns of HLA-C allotype recognition. J Immunol. 1998;161:571–577. [VSports手机版 - PubMed] [Google Scholar]
  • 23.Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science. 2002;296:1323–1326. doi: 10.1126/science.1070884. [DOI (VSports在线直播)] [PubMed] [Google Scholar]
  • 24.Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer cells. Nature. 2009;457:557–561. doi: 10.1038/nature07665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cooley S, McCullar V, Wangen R, et al. KIR reconstitution is altered by T cells in the graft and correlates with clinical outcomes after unrelated donor transplantation. Blood. 2005;106:4370–4376. doi: 10.1182/blood-2005-04-1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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. doi: 10.1182/blood-2011-04-347070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Foley B, Cooley S, Verneris MR, et al. Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C+ natural killer cells with potent function. Blood. 2012;119:2665–2674. doi: 10.1182/blood-2011-10-386995. ["V体育官网" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Burns LJ, Weisdorf DJ, DeFor TE, et al. IL-2-based immunotherapy after autologous transplantation for lymphoma and breast cancer induces immune activation and cytokine release: a phase I/II trial. Bone Marrow Transplant. 2003;32:177–186. doi: 10.1038/sj.bmt.1704086. [DOI] [PubMed] [Google Scholar]
  • 29.Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051–3057. doi: 10.1182/blood-2004-07-2974. [DOI] [PubMed] [Google Scholar]
  • 30.Geller MA, Cooley S, Judson PL, et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy. 2011;13:98–107. doi: 10.3109/14653249.2010.515582. [VSports - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bachanova V, Burns LJ, McKenna DH, et al. Allogeneic natural killer cells for refractory lymphoma. Cancer Immunol Immunother. 2010;59:1739–1744. doi: 10.1007/s00262-010-0896-z. ["VSports手机版" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gleason MK, Verneris MR, Todhunter DA, et al. Bispecific and trispecific killer cell engagers directly activate human NK cells through CD16 signaling and induce cytotoxicity and cytokine production. Mol Cancer Ther. 2012;11:2674–2684. doi: 10.1158/1535-7163.MCT-12-0692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194:23–28. doi: 10.1126/science.959840. [DOI] [PubMed] [Google Scholar]
  • 34.Wang L, Lawrence MS, Wan Y, et al. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N Engl J Med. 2011;365:2497–2506. doi: 10.1056/NEJMoa1109016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Campbell P, Pleasance E, Stephens P, et al. Subclonal phylogenetic structures in cancer revealed by ultra-deep sequencing. Proc Natl Acad Sci U S A. 2008;105:13081–13086. doi: 10.1073/pnas.0801523105. ["V体育平台登录" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481:506–510. doi: 10.1038/nature10738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Egan JB, Shi CX, Tembe W, et al. Whole-genome sequencing of multiple myeloma from diagnosis to plasma cell leukemia reveals genomic initiating events, evolution, and clonal tides. Blood. 2012;120:1060–1066. doi: 10.1182/blood-2012-01-405977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Snuderl M, Fazlollahi L, Le LP, et al. Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. Cancer Cell. 2011;20:810–817. doi: 10.1016/j.ccr.2011.11.005. [DOI (VSports在线直播)] [PubMed] [Google Scholar]
  • 39.Ene CI, Fine HA. Many tumors in one: a daunting therapeutic prospect. Cancer Cell. 2011;20:695–697. doi: 10.1016/j.ccr.2011.11.018. [V体育2025版 - DOI] [PubMed] [Google Scholar]
  • 40.Oravecz-Wilson KI, Philips ST, Yilmaz OH, et al. Persistence of leukemia-initiating cells in a conditional knockin model of an imatinib-responsive myeloproliferative disorder. Cancer Cell. 2009;16:137–148. doi: 10.1016/j.ccr.2009.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Clevers H. The cancer stem cell: premises, promises and challenges. Nat Med. 2011;17:313–319. doi: 10.1038/nm.2304. [DOI] [PubMed] [Google Scholar]
  • 42.Mullighan CG, Phillips LA, Su X, et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science. 2008;322:1377–1380. doi: 10.1126/science.1164266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Maley CC, Galipeau PC, Finley JC, et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat Genet. 2006;38:468–473. doi: 10.1038/ng1768. [VSports - DOI] [PubMed] [Google Scholar]
  • 44.Roche-Lestienne C, Soenen-Cornu V, Grardel-Duflos N, et al. Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment. Blood. 2002;100:1014–1018. doi: 10.1182/blood.v100.3.1014. [DOI (V体育安卓版)] [PubMed] [Google Scholar]
  • 45.Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nat Rev Cancer. 2012;12:335–348. doi: 10.1038/nrc3257. [DOI] [PubMed] [Google Scholar]
  • 46.Landau DA, Stojanov P, Lawrence MS, et al. Shifts in Intra-Clonal Dynamics Rather Than Novel Mutations Are the Main Engine Driving Tumor Evolution in Relapsed CLL. Blood. 2011;118:131–132. ["V体育官网入口" Google Scholar]
  • 47.Gatenby RA, Silva AS, Gillies RJ, Frieden BR. Adaptive therapy. Cancer Res. 2009;69:4894–4903. doi: 10.1158/0008-5472.CAN-08-3658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Dreger P, Dohner H, Ritgen M, et al. Allogeneic stem cell transplantation provides durable disease control in poor-risk chronic lymphocytic leukemia: long-term clinical and MRD results of the German CLL Study Group CLL3X trial. Blood. 2010;116:2438–2447. doi: 10.1182/blood-2010-03-275420. [VSports - DOI] [PubMed] [Google Scholar]
  • 49.Avigan D, Hari P, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Hsu KC. Proceedings from the National Cancer Institute’s Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: part II. Autologous Transplantation-novel agents and immunomodulatory strategies. Biol Blood Marrow Transplant. 2013 Dec;19(12):1661–9. doi: 10.1016/j.bbmt.2013.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.de Lima M, Porter DL, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Schmid C. Proceedings from the National Cancer Institute’s Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: Part III. Prevention and Treatment of Relapse after Allogeneic Transplantation. Biol Blood Marrow Transplant. 2014 Jan;20(1):4–13. doi: 10.1016/j.bbmt.2013.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES