Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jan 31.
Published in final edited form as: Nat Med. 2017 Jan 6;23(1):18–27. doi: 10.1038/nm.4241

T memory stem cells in health and disease

Luca Gattinoni 1, Daniel E Speiser 2, Mathias Lichterfeld 3, Chiara Bonini 4
PMCID: PMC6354775  NIHMSID: NIHMS1003141  PMID: 28060797

Abstract

T memory stem cells (TSCM) are a rare subset of memory lymphocytes endowed with the stem cell-like ability to self-renew and the multipotent capacity to reconstitute the entire spectrum of memory and effector subsets. Cumulative evidence in mice, non-human primates and humans indicates that TSCM are minimally differentiated cells at the apex of the hierarchical system of memory T lymphocytes. Here we describe emerging findings demonstrating that TSCM, owing to their extreme longevity and robust potential for immune reconstitution, are central players in many physiological and pathological human processes. We also discuss how TSCM stemness could be therapeutically leveraged to enhance the efficacy of vaccines and adoptive T-cell therapies for cancer and infectious diseases or, conversely, disrupted to treat TSCM-driven and sustained diseases such as autoimmunity, adult T-cell leukemia, and HIV-1.

Graphical Abstract

graphic file with name nihms-1003141-f0003.jpg


Immunological memory – the ability to remember and respond rapidly and more vigorously to subsequent encounters with a pathogen – has long been recognized in human history. The first documentation of immunological memory was provided by the Greek historian, Thucydides, who vividly described the plague that struck the city of Athens in 430 BC recounting that “this disease never took any man the second time”1. It took us more than two millennia to gain insights into the cellular basis of the immune system and to understand that immunological memory is a fundamental property of the adaptive immunity conveyed by B and T lymphocytes2.

Despite the enormous progress in our understanding of basic aspects of T-cell immunity, the ontogeny of memory T cells remains a matter of active debate3,4. It is clear, however, that immunological memory and protective immunity can last several decades and perhaps a lifetime even in the absence of re-exposure to the pathogen5,6. This astonishing stability of T-cell memory in spite of the high cellular turnover characterizing the immune responses and the lack of replenishment of antigen-specific T cells from hematopoietic stem cells (HSCs) due to constraints imposed by stochastic recombination of the T-cell receptor (TCR) and thymic involution, has sparked the idea that T-cell immunity could be maintained via stem cell-like memory T cells7. Over the past decade, the realization that memory T cells share a core transcriptional signature with HSCs8 and display functional properties found in stem cells, such as the capacity to divide asymmetrically to generate cellular heterogeneity9, has further strengthened the view that T cells, akin to all somatic tissues, may be hierarchically organized and sustained by antigen-specific T memory stem cells10.

In this Review, we outline emerging findings demonstrating that a subset of minimally differentiated memory T cells behave as antigen-specific adult stem cells. We also discuss recent evidence placing these T memory stem cells (TSCM) at center stage in many physiological and pathological human processes. Finally, we highlight ongoing efforts aiming either at harnessing the therapeutic potential of TSCM for adoptive immunotherapies or conversely at destabilizing the TSCM compartment to eliminate drug-resistant viral reservoirs or treat adult T-cell leukemia and autoimmune diseases. The conceptual work and key discoveries that have shaped this field of investigation are summarized in the Timeline.

The discovery of TSCM cells

Advances in multiparameter flow cytometry over the last 20 years have allowed us to dissect the heterogeneity of the T-cell compartment with ever-increasing precision11. In a seminal study, van Lier and colleagues identified human naïve, memory, and effector T-cell subsets based on the combinatorial expression of CD27 and CD45RA, with naïve cells expressing both molecules, whereas memory and effector cells expressing only CD27 or CD45RA, respectively12. Subsequent work by Sallusto et al.13 revealed the presence of two major functional subsets within the CD45RA memory T-cell pool: central memory T cells (TCM), which express the lymph node homing molecules CCR7 and CD62L and have limited effector functions, and CCR7CD62L effector memory T cells (TEM), which preferentially traffic to peripheral tissues and mediate rapid effector functions.

The idea that memory T cells may not solely be confined to the CD45RA T-cell compartment, but may also be present within what was considered to be an exclusive naïve T-cell realm, began to take shape following the identification in mice of a novel memory T-cell population characterized by a naïve-like phenotype but expressing high amounts of stem cell antigen-1 (SCA-1) and the memory markers interleukin-2 receptor β (IL-2Rβ) and chemokine C-X-C motif receptor 3 (CXCR3)14. These cells were termed TSCM based on the observation that they were capable of sustaining graft-versus-host disease (GVHD) upon serial transplantation in allogeneic hosts and that they could reconstitute the full diversity of memory and effector T-cell subsets while maintaining their own pool size through self-renewal14. Identifying the human counterpart of TSCM, however, has not been straightforward mainly due to the lack of a human ortholog of SCA-1, the prototypical marker of mouse TSCM. Though it was known that a significant fraction of long-lived antigen-specific CD8+ and CD4+ memory T cells displayed a naïve-like phenotype (CD45RA+CCR7+CD27+) years after infection with EBV15 or vaccination with attenuated smallpox or yellow fever (YF) viruses16,17, a precise set of surface markers to pinpoint this elusive memory phenotype in humans was missing. The breakthrough came with the demonstration that mouse TSCM could be successfully generated in vitro from naïve precursors by activating the WNT-β-catenin signaling pathway using the WNT ligand, WNT3A, or inhibitors of glycogen synthase kinase-3β18. Translated to humans, this strategy has recently allowed the identification of human TSCM19. Similar to their murine counterparts, human and non-human primate (NHP) TSCM are clonally expanded cells expressing a largely naïve-like phenotype in conjunction with a core of memory markers such as CD95, CXCR3, IL-2Rβ, CD58 and CD11a19,20. These cells represent a small fraction of circulating T lymphocytes (≈ 2–3%). Interestingly, the frequency of circulating TSCM does not significantly vary with age21, but it appears to be heritable and associated with single nucleotide polymorphisms at a genetic locus containing CD9522, suggesting a potential role of FAS signaling in the regulation of TSCM homeostasis. TSCM exhibit all the defining properties of memory cells, including a diluted content of TCR excision circles, the ability to rapidly proliferate and release inflammatory cytokines in response to antigen re-exposure, and a dependence on IL-15 and IL-7 for homeostatic turnover19,23. Despite being functionally distinct from naïve T cells, they share similar recirculation patterns and distribution in vivo as evidenced by detailed compartmentalization studies in NHP24. For instance, TSCM are found more abundantly in lymph nodes compared to the spleen and bone marrow and are virtually absent from peripheral mucosae24. Thus, TSCM represent a subset of minimally differentiated T cells characterized by phenotypic and functional properties bridging naïve and conventional memory cells (Fig. 1).

Figure 1: Hierarchical model of human T-cell differentiation.

Figure 1:

Following antigen priming, naïve T cells (TN) progressively differentiate into diverse memory T-cell subpopulations and ultimately into terminally differentiated effector T cells (TTE). T-cell subsets are distinguished by the combinatorial expression of the indicated surface markers. As TN progressively differentiate into TTE, they lose or acquire specific functional and metabolic attributes. TSCM, T memory stem cell; TCM, central memory T cell; TEM, effector memory T cell; ΔΨm, mitochondrial membrane potential.

TSCM cells: Evidence of stemness

The concept of stemness embraces the capacity both to self-renew and to generate the entire spectrum of more differentiated cells25. When the existence of a stem cell pool of memory T lymphocytes was initially postulated by Fearon and colleagues7, the authors pointed to TCM as putative T memory stem cells. This assumption was based on the evidence that TCM are less differentiated than TEM and effector cells, as shown by their longer telomeres and lower expression of perforin, granzymes and other effector molecules13. Furthermore, it was intuitive to assume that the pool of T memory stem cells should be confined in lymph nodes and secondary lymphoid organs, and TCM were, at that time, the only antigen-experienced T cells known to express CCR7 and CD62L. The notion that TCM might function as T memory stem cells was further supported by subsequent findings demonstrating that TCM have superior immune reconstitution capacity and a greater ability to persist in vivo than TEM26. Recent clonogenic experiments in mice based on single cell serial transfer have formally demonstrated the ability of murine TCM to self-renew and generate TEM and effector progenies in vivo27,28. Strikingly, TEM were unable to serially reconstitute the host even when transferred at 100-fold higher numbers, showing a limited capacity for self-renewal. Although these experiments did not evaluate TSCM, these results, combined with that of sophisticated experiments tracking T cell fates in mice based on genetic barcoding29 and on single naïve T-cell transfer30, provided strong support for the progressive model of T-cell differentiation originally developed by Sallusto and Lanzavecchia31. Indeed, three separate models have been proposed to explain memory T-cell differentiation3: according to the first 2 models memory T cells originate from effectors either after26 or before32 the peak of T-cell expansion. The progressive differentiation model, on the contrary, suggests that memory T cells are derived directly from naïve lymphocytes upon priming, and further differentiate into shorter-lived effector subsets in a hierarchical differentiation tree, similar to that of other organ systems31 (Fig. 1). Using hematopoietic stem cell transplantation (HSCT) from haploidentical donors as a model system to study T-cell differentiation, two independent groups have recently showed at polyclonal, antigen-specific, and clonal levels, that human TSCM differentiate directly from naïve precursors, emerging early upon in vivo priming33,34. By multiparametric flow cytometry and TCR sequencing of individual T lymphocytes it was possible to trace and quantify, at the clonal level, the in vivo differentiation landscapes of transferred naïve and antigen-experienced T cells, highlighting TSCM as privileged players in the diversification of naïve cells upon priming33,34. Indeed, discrete T-cell subsets traced across HSCT behaved preferentially within a progressive framework of differentiation. Notably, only naïve T cells and TSCM were able to reconstitute the entire heterogeneity of memory T-cell subsets, including TSCM33. A fraction of originally TEM reverted to a TCM phenotype33. By contrast, only a very limited number of TCM and TEM converted to TSCM33. Echoing these findings, the transfer of genetically-modified virus-specific T cells reconstituted the full diversity of the T-cell memory compartment – inclusive of TSCM, TCM and TEM – only when TSCM were present within the infused cell product35. Altogether these results strengthen earlier in vitro observations in human19 and NHP24 showing that the potential to form diverse progeny is progressively restricted proceeding from TSCM to TCM and TEM. Thus, granting some level of plasticity to the system, these data point to a progressive model of T-cell differentiation, in which TSCM are at the apex of the hierarchical tree. In line with this concept, the gene expression profile of human T-cell subsets partitions TSCM with antigen-experienced T cells, and places them at a hierarchically superior level than TCM19,23,36,37.

The concept of stemness also involves self-renewal and implicates long-term persistence25. The long-term persisting ability of TSCM and other antigen-experienced T cells cannot be easily addressed in humans, since naïve T cells are continuously generated, and several antigenic contacts might occur after the initial encounter. Longitudinal monitoring of genetically-engineered lymphocytes infused as antigen-experienced cells and distinguishable from endogenous lymphocytes thanks to the retroviral integration and transgene expression, has recently allowed the tracking of single T-cell clonotypes over time. In patients affected by ADA-SCID, genetically-engineered TSCM persisted and preserved their precursor potential for decades38. In leukemic patients treated with haploidentical HSCT and donor lymphocytes retrovirally transduced to express a suicide gene, engineered lymphocytes were traced for up to 14 years35. This study revealed that the extent of expansion and the amount of persisting gene-marked T cells tightly correlate with the number of TSCM infused, indicating that this subset of memory cells is endowed with enhanced proliferative potential, immune reconstitution capacity and longevity35. Interestingly, the same observation has been reported in a clinical trial based on the infusion of autologous T cells genetically-engineered to express a chimeric antigen receptor (CAR)39, underscoring that this phenomenon is not confined to the HSCT model. In patients treated with suicide gene therapy, by combining T-cell sorting with sequencing of integration, TCRα and TCRβ clonal markers, it was possible to show that dominant long-term T-cell clones preferentially originate from infused TSCM and to a lesser degree from TCM clones35. Together, these results indicate that human TSCM have an exceptional capacity to persist long-term. Similar conclusions were reached by monitoring T-cell subset dynamics in NHP infection models24 and HIV-1 patients undergoing antiretroviral therapy (ART)40, in which antigen load and time of antigen exposure can be precisely controlled. Taking advantage of the peculiar biology of the Tat-specific epitope TL8, which uniformly undergoes escape mutation within 4–5 weeks after Simian Immunodeficiency Virus (SIV) infection, Lugli et al.24 investigated the persistence of different memory T-cell subsets in the virtual absence of any antigen perturbation. In this setting, TSCM were able to persist at unchanged levels for up to 70 days after infection, whereas TCM and TEM contracted 10-fold and 100-fold, repesctively24. Likewise, pharmacological antigen withdrawal in ART-treated HIV-1 patients was associated with a decline of HIV-1-specific TEM and terminally differentiated effector cells (TTE), whereas TSCM were restored and even expanded under these conditions40. Mirroring these findings, YF-specific T-cell subsets declined after vaccination with attrition rates progressively increasing along with a differentiation from TCM to TEM and TTE36. Remarkably, the frequency of YF-specific TSCM was stably maintained even 25 years after vaccination36. Taken together, this series of studies provides compelling evidence that human TSCM are generated directly from naïve lymphocytes and are endowed with long-term self-renewal capacity and multipotency.

TSCM cells in antimicrobial immune defense and after vaccination

Human TSCM have been increasingly identified in acute and chronic infections caused by a variety of pathogens, including viruses, bacteria and parasites19,35,36,4042. These results demonstrate that TSCM are commonly generated during natural immune responses against foreign pathogens, but the underlying mechanisms remain poorly understood. Human studies are limited in that the exact time of infection is usually unknown, making it difficult to study T-cell priming and kinetics. By contrast, active vaccination offers the possibility to induce an immune response in a supervised fashion. Smallpox and YF vaccines are particularly suitable models of human primary acute viral infection as they consist of live attenuated, replication competent viruses capable of inducing strong immune responses with consequent clinical symptoms43. Using YF vaccination as model system, the kinetics of TSCM formation and long-term maintenance have recently been studied in great detail36. Consistent with findings from studies of SIV infection in NHP24, YF-specific TSCM were detectable at early time points after vaccination when the immune response was dominated by effector T cells36. These TSCM persisted at stable levels, becoming the major YF-specific memory T-cell population in the circulation decades after the initial immunization36. Considering that YF vaccination provides life-long protection43, it is reasonable to assume that TSCM play a central role in the maintenance of long-term T-cell memory.

The presence of a relevant pool of TSCM might also be essential for the control of persisting infections, in which effector T cells undergoing functional exhaustion and replicative senescence need to be continuously replenished by less-differentiated T-cell subsets4446. Interestingly, recent studies in chronic viral40,47 and parasitic infections42 have shown the existence of a negative correlation between the severity of disease and the frequency of circulating TSCM. It is unclear whether these observations result from the inability of TSCM to be maintained under conditions of strong inflammation and high antigenic load, or vice versa, the lack of physiological numbers of TSCM impairs the ability of the immune system to keep the infection in check. However, emerging findings suggest that TSCM are a fundamental pillar of immune homeostasis as high levels of infection and subsequent functional perturbation of the TSCM compartment have been linked to the development of symptomatic immune deficiency following SIV and HIV-1 infections48,49. Indeed, high quantities of SIV DNA were found in CD4+ TSCM from rhesus macaques, who typically develop an AIDS-like clinical picture when left untreated, but not in CD4+ TSCM from SIV-infected sooty mangabeys, a group of NHP who are refractory to clinical or laboratory signs of immune deficiency even when high levels of virus circulate in the peripheral blood48,50,51. Resonating with this observation, viremic non-progressors – a rare group of untreated HIV-1 patients, who develop high levels of HIV-1 replication in the absence of clinical immune deficiency – exhibit reduced levels of HIV-1 DNA in CD4+ TSCM in comparison to HIV-1 patients with ordinary rates of disease progression, who instead show high amounts of HIV-1 infected TSCM despite a relative depletion of the total CD4+ TSCM compartment49. Altogether these results underscore a critical function of TSCM in sustaining long-lasting cellular immunity against acute and chronic microbial infections.

Given the pivotal role of TSCM in maintaining life-long immunological memory, it would be desirable to develop vaccines capable of inducing significant numbers of TSCM. The majority of clinical vaccine formulations designed to stimulate CD8+ T-cell-mediated immunity induce predominantly TEM, with only little emergence of memory cells52,53. These vaccines are generally poorly efficient and rarely protective compared to those inducing protective antibodies2,54. Indeed, current T-cell vaccines appear unable to trigger mechanisms that are key for the development of memory T cells, including optimal signaling via the TCR and induction of appropriate metabolic programs, transcription factors, and chromatin reorganization55. Considering that the activation of CD8+ T cells under conditions of low-level inflammation enhances memory cell formation, one may consider that novel vaccines should preferentially activate T cells without triggering the excessive release of pro-inflammatory cytokines56. It is however debatable whether optimal generation of memory T cells requires the avoidance of effector cell differentiation. This is illustrated by the fact that natural infections generate sound memory T-cell responses, including TSCM, despite initial predominance of effector cells43. Much work remains to be done in this area, however, the induction of TSCM by novel vaccines should not be at the expense of more differentiated TEM and tissue-resident memory cells, which assure immediate protection at the entry site of re-infection in peripheral tissues5759. Ideally, new vaccines will be able to recreate the large heterogeneity of memory cells, including long-lived TSCM, that human pathogens and their pathophysiological properties induce in vivo60,61.

TSCM cells in human diseases

The complex biology of TSCM can make it difficult to discriminate between their protective and pathogenic effects because the very characteristics that enable TSCM to represent the backbone of life-long cellular immunity under physiologic conditions may empower these cells to drive disease pathogenesis62. This seems particularly relevant in the setting of a growing list of immune-mediated diseases associated with aberrant and autoreactive memory T cells. For instance, recent correlative studies have suggested an increased frequency and activation state of CD8+ TSCM in individuals with aplastic anemia, a disease mediated by autoreactive cytotoxic T cells targeting hematopoietic progenitors63. Moreover, an elevated number of CD8+ TSCM after immunosuppressive treatment was associated with treatment failure and subsequent disease relapse63. Elevated quantities of TSCM were also noted in patients with uveitis, but not with systemic lupus erythematosus, an immune-mediated disease primarily characterized by autoreactive humoral responses63. Further pointing towards a role of TSCM in the pathogenesis of autoimmune diseases and other illnesses of the lymphatic system, a recent genome-wide association study found a strong association between genetic polymorphisms affecting susceptibility to juvenile idiopathic arthritis or chronic lymphocytic leukemia, and the frequency of CD4+ TSCM22. How TSCM can influence autoimmune diseases will have to be studied in dedicated investigations, but based on current knowledge it is reasonable to hypothesize that long-lasting autoreactive or abnormally activated TSCM may induce self-renewing inflammatory cellular responses that are responsible for the durable, and in most cases life-long persistence of such diseases64. The possible role of TSCM in other diseases with profound disturbance of cellular immune responses, such as autoimmune hepatitis, thyroiditis, type I diabetes and certain types of glomerulonephritis, are currently unknown but represent a high priority area of future research.

In addition to their role in autoimmunity, TSCM may have a distinct role in viral diseases in which T cells represent the predominant targets, such as infections caused by CD4+ T-cell tropic retroviruses. Notably, work in the context of HIV-1 infection has shown that CD4+ TSCM can effectively support both productive viral replication and a transcriptionally-silent form of infection65. Moreover, by infecting long-lived CD4+ TSCM, HIV-1 is able to exploit their stemness to establish an extremely durable, self-renewing viral reservoir that can persist for decades despite ART, and continuously replenish virally-infected cells, perpetuating a disease they are meant to restrict66. Indeed, the half-life of HIV-1-infected TSCM in ART-treated individuals has been estimated to last for 277 months, a time period significantly longer than that observed for viral reservoirs established in more short-lived T-cell populations67. In line with these observations, phylogenetic studies demonstrated close associations between viruses circulating early after HIV-1 infection and viral sequences isolated from CD4+ TSCM after almost a decade of suppressive ART66. Notably, the ability to use CD4+ TSCM as a long-term viral reservoir also seems to occur in individuals infected with HTLV-1, a retrovirus related to HIV-1 that is the primary cause of adult T-cell leukemia (ATL). Emerging data indicate that transformed, HTLV-1 infected CD4+ TSCM can act as progenitors for dominant circulating ATL clones, and efficiently repopulate ATL clones upon transplantation in animal models68 suggesting that they can serve as a cancer stem cell population responsible for propagating and maintaining HTLV-1 infected malignant cells.

Targeting TSCM cells for therapy

Harnessing TSCM cells for adoptive T-cell therapy

The extreme longevity, the robust proliferative potential and the capacity to reconstitute a wide-ranging diversity of the T-cell compartment make TSCM an ideal cell population to employ in adoptive immunotherapy (Fig. 2). Driven by the growing success of clinical trials based on the transfer of naturally occurring and genetically-engineered tumor-reactive T lymphocytes, adoptive immunotherapies are rapidly becoming a real therapeutic option for patients with cancer69,70. Although these regimens can induce complete and durable tumor regressions in patients with advanced cancer, current response rates remain mostly inadequate underscoring the need for further improvements69,70. There is now extensive evidence indicating that objective responses strongly correlate with the level of T-cell engraftment and peak of expansion earlier after transfer7177. T-cell persistence, though not strictly indispensable in certain conditions7275,78, has also been associated with the likelihood of objective responses in numerous trials 76,77,7983 and might be required to sustain durable remissions84. These parameters are considerably influenced by the composition of the infused T-cell product as T-cell subsets differ widely in terms of proliferative capacity, immune reconstitution and long-term survival10,85. Indeed, the administration of cells with longer telomeres81,86 or cell products comprising higher fractions of CD62L+, CD28+ or CD27+ T cells has been shown to correlate with objective tumor responses in patients81,8688, suggesting that less-differentiated T cells are therapeutically superior to TTE. Notably, the engraftment and expansion of T cells engineered to express a CD19-specific CAR39 or a suicide gene35 correlated with the frequency of infused CD8+CD45RA+CCR7+ TSCM. Adoptive transfer experiments in mice using defined T-cell subsets have formally demonstrated that the infusion of less-differentiated CD62L+ T-cell populations results in enhanced T-cell engraftment, expansion and persistence, ultimately leading to more profound and durable tumor regressions18,19,8993. Consistent with the developmental hierarchy, minimally differentiated TSCM mediate more potent antitumor responses than TCM, which in turn are more effective than highly differentiated TEM18,19,94. Some level of plasticity, however, must be granted to the hierarchical model of memory T-cell differentiation. In NHP, genetically-engineered CMV-specific effectors derived from purified TCM proved superior to effectors derived from TEM in terms of in vivo expansion and persistence, showing that even after in vitro manipulation and apparently a similar degree of terminal differentiation, T cells maintain some characteristics of the subset of origin, and can possibly, at least in part, revert to that original phenotype and function95.

Figure 2: TSCM-based therapeutic interventions for human diseases.

Figure 2:

T memory stem cells (TSCM) can be either tamed (left panel) to treat TSCM-driven diseases such as autoimmunity, T-cell leukemia and T-cell tropic infections or exploited (right panel) to potentiate T cell-based immunotherapies against cancer and infectious diseases. Left panel: WNT antagonists or short hairpin RNA (shRNA) targeting key molecules involved in WNT signaling such as T cell factor 7 (TCF7) could be used to disrupt long-lasting, self-renewing TSCM reservoirs by driving them to differentiate into short-lived subsets such as effector memory T cells (TEM). Nanoparticle or aptamer technology could be employed to specifically target CD4+ T cells or virally-infected T cells. Right panel: patient- or donor-derived naïve-like T cells can be used to generate and in vitro expand TSCM with or without gene engineering. Gene modifications include the insertion of tumor or virus-specific chimeric antigen receptor (CAR) or T cell receptor (TCR) genes, tumor or virus-specific TCR gene editing, suicide gene transfer in the context of donor lymphocyte infusion following hematopoietic stem cell transplantation, and CCR5 deletion in the setting of HIV-1 infection. Virus-specific TSCM can also be expanded from the naturally-occurring antigen-specific TCR repertoire through in vitro sensitization protocols favoring the generation of TSCM. TN, naïve T cell; TCM, central memory T cell; APC, antigen presenting cell.

Despite overwhelming preclinical data indicating a therapeutic advantage to transferring tumor-reactive CD62L+ T-cell subsets18,19,8993, clinical trials have largely employed unselected intratumoral or peripheral blood mononuclear cell (PBMC)-derived T-cell populations. Tumor infiltrating lymphocytes are typically in a state of terminal differentiation and functional exhaustion making the isolation of early memory T-cell subsets impractical96,97. However, the selection of less-differentiated T-cell subsets becomes realistic and desirable in the context of immunotherapies aiming at conferring tumor reactivity to circulating T cells via TCR or CAR gene-engineering. The isolation of less-differentiated T-cell populations also has the advantage of reproducibly generating more consistent and defined T-cell products. Indeed, PBMC composition can vary significantly between individuals as a consequence of age98, pathogen exposure99, and prior systemic treatments100. Moreover, unselected populations containing high proportions of TEM and effector cells might fail to generate viable clinical products due to poor in vitro cell expansion101. Recently, two clinical trials in which CD19-specific CAR T cells were generated from isolated TCM have been reported84,102,103. This strategy led to the generation of infusion products comprising significantly more TEM than those originating from unselected PBMC indicating that, in the absence of culture conditions restraining T-cell differentiation18,104108, the benefit of depleting highly-differentiated T cell subsets is outweighed by the concomitant removal of naïve and TSCM102. Notwithstanding the reduction of less-differentiated T-cell subsets, the rates of objective remissions in acute lymphoblastic leukemia (ALL) patients were comparable to trials using unselected T-cell populations72,73,76,102,109,110. Whether differences in manufacturing and T-cell product composition will affect rates and duration of clinical responses in other diseases and settings remains to be shown.

So far, the clinical exploitation of TSCM has been hindered by their relative paucity in the circulation19,20 and the lack – until recently – of robust, clinical-grade manufacturing protocols capable of generating and maintaining this cell type in vitro. These strategies rely on programming and redirecting TSCM from naïve-like T cells isolated from PBMC23,111 (Fig. 2). Although the isolation of naïve T cells adds complexity to the manufacturing process, it is a critical step because the presence of more differentiated T-cell subsets during naïve T-cell stimulation accelerates naïve T-cell differentiation into TEM and TTE cells112. It should also be considered that purifying large numbers of specific cell subsets over multiple parameters under GMP conditions is becoming increasingly accessible thanks to recent developments in clinical cell sorting technologies85,113. IL-7 and IL-15 have successfully been used to generate tumor-redirected or suicide gene-modified TSCM from naïve cell precursors23 (Fig. 2). IL-7 is essential for the development of these cells23,114, while IL-15 primarily sustains their expansion23. IL-7 and IL-15-programmed TSCM possess a core gene signature of naturally occurring TSCM, display an enhanced proliferative capacity compared to other T-cell subsets and are uniquely capable of expanding and mediating GVHD upon serial transplantation23. This cytokine combination could also be employed to generate large numbers of TCR gene-edited TSCM by combining Zinc Finger Nuclease sets specific for the endogenous TCR gene loci with lentiviral vectors encoding tumor-specific TCRs115 (Fig. 2). Moreover, the ability of IL-7 and IL-15 to support the formation and expansion of TSCM makes it an ideal strategy to generate TSCM without the need to redirect their specificity. This may be particularly suitable for the generation of virus-specific TSCM for the treatment and prevention of life-threatening infections after transplantation (Fig. 2) as infection control can be obtained by transferring relatively small numbers of virus-specific memory cells116. A demonstration that IL-7 and IL-15 could be successfully employed to generate and expand virus-specific TSCM starting from isolated naïve-like cells was recently provided by Volk and colleagues117. This protocol could also be adapted to generate CAR-modified virus-specific TSCM, which may lower the risk of GVHD given the restricted TCR repertoire and exhibit additional proliferative and survival advantages as result of the in vivo triggering of the native virus-specific TCRs by antigens from persistent viruses80,118. Another clinical-grade strategy promoting the generation of tumor-reactive TSCM is based on the activation of naïve-like lymphocytes in the presence of IL-7, IL-21 and the WNT agonist TWS119111. Although both IL-15119,120 and IL-21121-123 have been implicated in the generation and maintenance of memory T cells, IL-21 is more effective in restraining T-cell differentiation105 due to its specific ability to activate STAT3 signaling124 and to sustain the expression of WNT-β-catenin transcription factors TCF7 and LEF1105. TWS119 provides a synergistic effect with IL-21 to induce maximal expression of TCF7 and LEF1 by stabilizing β-catenin111. CAR-modified TSCM generated under these culture conditions are phenotypically, functionally, and transcriptionally equivalent to their naturally occurring counterparts111. Moreover, they exhibit metabolic features characteristic of long-lived memory T cells such as a high spare respiratory capacity125 and low glycolytic metabolism126. Although these culture conditions profoundly inhibit T-cell proliferation, TSCM can be efficiently redirected against a tumor antigen and expanded to clinically relevant numbers111. More importantly, CAR-modified CD8+ TSCM mediated superior and more durable anti-tumor responses than cells generated with protocols currently employed in clinical trials111. CAR-modified TSCM may also provide an attractive approach for immunotherapy in the setting of non-malignant diseases, such as HIV-1 infection or other chronic viral illnesses127,128 (Fig. 2). Altogether, these studies provide both a strong scientific rationale and practical methodologies for the rapid advancement of TSCM in human clinical trials of adoptive immunotherapy129.

Disrupting TSCM cell reservoirs in retroviral infections and autoimmune diseases

The emerging role of CD4+ TSCM in the pathogenesis of chronic viral infections such as HIV-1 and HTLV-1 infection may also offer novel opportunities to prevent, treat or cure these diseases. In the context of HIV-1 infection, specific interventions that eliminate HIV-1-infected CD4+ TSCM may allow for the destabilization of HIV-1 reservoirs by reducing the number of HIV-1-infected source cells from which new HIV-1+ viral and cellular progeny can continuously originate, despite suppressive ART. As the molecular programs governing the stem cell-like behavior of TSCM continue to be understood, new molecules regulating proliferation and self-renewal of TSCM may represent attractive targets for reducing viral persistence in CD4+ TSCM. For instance, WNT-β-catenin signaling has been identified as a key driver for the homeostasis of TSCM18, and pharmaceutical inhibition of this pathway may therefore translate into a more limited ability of HIV-1 to use the TSCM compartment for maintaining survival of virally-infected cells (Fig. 2). This approach may be facilitated by the availability of existing pharmacological inhibitors of WNT-β-catenin designed to target cancer stem cells130. Although such as strategy might be not entirely specific for eliminating HIV-1-specific CD4+ TSCM, advances in nanotechnology may allow for selective delivery of WNT-β-catenin antagonists or short hairpin RNAs targeting key mediators of WNT signaling to CD4+ T cells or virally-infected cells via nanoparticles or aptamer-based targeting systems131,132 (Fig. 2). Similar strategies are also conceivable to target HTLV-1-infected TSCM in the setting of ATL or to disrupt long-lasting reservoirs of autoreactive TSCM in autoimmune diseases. Additionally, recent advances in ex-vivo gene editing may allow the design of CD4+ TSCM that are intrinsically resistant to HIV-1, through e. g. targeted deletion of the chemokine receptor CCR5, which is necessary for viral entry133, thus mimicking the CCR5Δ32 mutation known to confer resistance to HIV-1 infection134 (Fig. 2). Such a population of long-lasting, HIV-1-resistant CD4+ T cells could be used in adoptive immunotherapy strategies to establish a durable cellular immune system that is no longer able to support HIV-1 infection, and may allow for drug-free remission of HIV-1 infection.

Concluding Remarks

TSCM are rare antigen-experienced T cells, likely generated directly from naïve lymphocytes and endowed with long-term self-renewal capacity and multipotency. Compelling evidence in mice, NHP, and humans points towards a scenario in which TSCM represent the apex of the memory T-cell differentiation tree. Their longevity and their capacity to reconstitute the entire heterogeneity of the T-cell memory compartment entail a double edged – protective or pathogenic – role for TSCM in human diseases. The increasingly recognized protective role of TSCM in acute and chronic infections makes them optimal candidates for therapeutic exploitation in vaccination and adoptive T-cell therapy against infectious diseases and cancer. Conversely, their relevance in the pathogenesis of autoimmunity, adult T-cell leukemia and HIV-1, makes TSCM an attractive target to tame for these pathological conditions. Several issues regarding TSCM biology remain to be addressed: characterization of their metabolic requirements, epigenetic and transcriptional programs, and anatomical niches (Box 1) will possibly guide innovative TSCM-based therapeutic interventions for human diseases.

Box 1: TSCM cell biology: outstanding questions.

Several questions regarding TSCM biology have yet to be addressed. A major unresolved issue is how TSCM physiologically form during the course of an infection and the impact of the strenght of antigen stimulation. Is TSCM fate immediately programmed at the time of naïve T cell priming or is it shaped throughout the number of antigen encounters and the diverse inflammatory environments that their progenies experience in the acute phase of the infection? A glimpse into TSCM transcriptional and epigenetic landscapes19,36,37,135,136, and early work exploring TSCM metabolism111,135 have begun to shed light on the molecular and metabolic programs regulating TSCM formation and homeostasis, but much ground remains to be covered. For instance, it is still unknown whether and to what extent asymmetric partitioning of key transcription factors137,138 and metabolic master regulators139,140 is programming TSCM formation. Additionally, studies examining TSCM anatomical niches are entirely missing. Progress in these area of investigation has been hampered to some extent by the rareness of TSCM in the circulation, which is a limiting factor for epigenetic, proteomic and metabolomic studies. A major hurdle is the lack of mouse infection models capable of generating robust numbers of TSCM, which so far has precluded researchers to precisely evaluate specific gene contributions to TSCM physiology and physiopathology with genetic tools, and to image TSCM dynamics in tissues by real-time in vivo microscopy.

TSCM epigenetic and transcriptional programs.

Genome-wide transcriptomic analyses of whole19,135 and yellow fever-specific TSCM36,37 have shown a high relatedness between these cells and central memory T cells (TCM). These findings suggest that the majority of signaling pathways and transcriptional factors that shape the development and maintenance of TCM might be to a certain degree involved in the regulation of TSCM. This reasoning is exemplified by the demonstration that WNT-β-catenin signaling, which is essential for TCM formation and long-term survival141144, is also critical for the generation of TSCM18,19,111. Likewise, tempering mTOR signaling has been shown to enhance the development of both TCM145,146 and TSCM135. Whether specific transcriptional networks are uniquely activated to influence TSCM fate is unknown. It is also currently unclear what is the role of CD95-FASL signaling in TSCM homeostasis. The epigenetic programs in TSCM remain largely undefined. Emerging genome-wide analysis of histone methylation on two histone H3 lysine residues (H3K4me3 and H3K27me3) in naïve and in vitro-generated murine CD8+ T cell subsets have revealed that chromatin accessibility is mostly regulated in a progressive fashion further supporting a hierarchical model of T-cell differentiation, in which TSCM represent the least differentiated antigen-experienced T cell subset136.

TSCM metabolism.

A growing body of work has recently highlighted the importance of cell metabolism in regulating the activity and fate commitment of T lymphocytes147. Fatty acid oxidation, increased mitochondrial biomass and spare respiratory capacity (SRC) have been shown to support the development of memory T cells and confer on them a bioenergetic advantage necessary to sustain rapid recall responses125,148. Conversely, aerobic glycolysis has been associated with the formation of short-lived terminally differentiated effector cells and defects in the establishment of T cell memory126. Emerging findings indicate that naturally-occurring and in vitro-generated human TSCM also exhibit the prototypical ‘metabolic signature’ of memory cells with reduced glycolytic flux, preferential lipid oxidative metabolism, and high SRC111,135. Recently, HSC and T cell stemness have been linked to presence of decreased mitochondrial membrane potential (ΔΨm)149. Consistent with these findings, TSCM display lower ΔΨm than other antigen-experienced T cell subsets, including TCM135,149. Whether TSCM maintain fused mitochondrial networks with tight cristae organization, which have been shown to facilitate electron transport chain activity in conventional memory T cells150 remains to be determined. Future area of research also include a more global characterization of the TSCM metabolome and a deeper understanding of the role of amino acids in TSCM metabolism and function.

TSCM anatomical niches.

Stem cell niches are instrumental in regulating stem cell behavior and tissue homeostasis151. Specialized niches in the bone marrow provide not only physical support but also soluble factors and cellular interactions that guide HSCs’ decision to either self-renew or differentiate151. Increasing evidence has recently underscored the critical role of the bone marrow also in sustaining life-long persistence of conventional memory T cells152155. Whether the bone marrow can similarly serve as a niche for TSCM is a fundamental question that needs to be addressed. Alternatively, it might be evaluated whether, akin to naïve T cells156, TSCM rely on homeostatic cues provided by fibroblastic reticular cell niches within T-cell zones of secondary lymphoid organs. Finally, it would be critical to characterize cell contact-dependent cross-talk, cytokine networks and metabolite constituents regulating TSCM differentiation and function in their niches.

Acknowledgments

Funding: This work was supported by the Intramural Research Program of the US National Institutes of Health, National Cancer Institute, Center for Cancer Research (ZIABC011480), the 2014 US National Institutes of Health (NIH) Bench-to-Bedside Award, the NIH grants AI098487, AI106468, AI114235, AI117841, AI120008, AI124776, the Cancer Research Institute (NY), the Ludwig Cancer Research (NY), the Swiss Cancer League (3507–08-2014), the Swiss National Science Foundation (320030_152856, CRSII3_160708), the SwissTransMed (KIP 18), the Italian Association for Cancer Research and the SUPERSIST (EU-FP7 project).

Timeline: T-cell stemness and TSCM: milestones and key discoveries

TSCM

T memory stem cells

GVHD

graft versus host disease

HIV-1

Human Immunodeficiency Virus-type 1

SIV

Simian Immunodeficiency Virus

HTLV-1

, Human T-cell lymphotropic virus type 1

CAR

Chimeric antigen receptor

Footnotes

Disclosure of Conflicts of interest:

All authors declare that they have no competing interests.

“δὶς γὰρ τὸν αὐτόν, ὥστε καὶ κτείνειν, οὐκ ἐπελάμβανεν.”

“For this disease never took any man the second time so as to be mortal.”

Thucydides, The History of the Peloponnesian War.

Translation by Thomas Hobbes

References

  • 1.Thucydides & Hobbes T Peloponnesian Warre, (Charles Harper, London, 1676). [Google Scholar]
  • 2.Sallusto F, Lanzavecchia A, Araki K & Ahmed R From vaccines to memory and back. Immunity 33, 451–463 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ahmed R, Bevan MJ, Reiner SL & Fearon DT The precursors of memory: models and controversies. Nature reviews. Immunology 9, 662–668 (2009). [DOI] [PubMed] [Google Scholar]
  • 4.Restifo NP & Gattinoni L Lineage relationship of effector and memory T cells. Current opinion in immunology (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Demkowicz WE Jr., Littaua RA, Wang J & Ennis FA Human cytotoxic T-cell memory: long-lived responses to vaccinia virus. J Virol 70, 2627–2631 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hammarlund E, et al. Duration of antiviral immunity after smallpox vaccination. Nature medicine 9, 1131–1137 (2003). [DOI] [PubMed] [Google Scholar]
  • 7.Fearon DT, Manders P & Wagner SD Arrested differentiation, the self-renewing memory lymphocyte, and vaccination. Science 293, 248–250 (2001). [DOI] [PubMed] [Google Scholar]
  • 8.Luckey CJ, et al. Memory T and memory B cells share a transcriptional program of self-renewal with long-term hematopoietic stem cells. Proceedings of the National Academy of Sciences of the United States of America 103, 3304–3309 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ciocca ML, Barnett BE, Burkhardt JK, Chang JT & Reiner SL Cutting edge: Asymmetric memory T cell division in response to rechallenge. J Immunol 188, 4145–4148 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gattinoni L, Klebanoff CA & Restifo NP Paths to stemness: building the ultimate antitumour T cell. Nature reviews. Cancer 12, 671–684 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chattopadhyay PK, Gierahn TM, Roederer M & Love JC Single-cell technologies for monitoring immune systems. Nature immunology 15, 128–135 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hamann D, et al. Phenotypic and functional separation of memory and effector human CD8+ T cells. The Journal of experimental medicine 186, 1407–1418 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sallusto F, Lenig D, Forster R, Lipp M & Lanzavecchia A Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708–712 (1999). [DOI] [PubMed] [Google Scholar]
  • 14.Zhang Y, Joe G, Hexner E, Zhu J & Emerson SG Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nature medicine 11, 1299–1305 (2005). [DOI] [PubMed] [Google Scholar]
  • 15.Long HM, et al. MHC II tetramers visualize human CD4+ T cell responses to Epstein-Barr virus infection and demonstrate atypical kinetics of the nuclear antigen EBNA1 response. The Journal of experimental medicine 210, 933–949 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Miller JD, et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity 28, 710–722 (2008). [DOI] [PubMed] [Google Scholar]
  • 17.Akondy RS, et al. The yellow fever virus vaccine induces a broad and polyfunctional human memory CD8+ T cell response. J Immunol 183, 7919–7930 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gattinoni L, et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nature medicine 15, 808–813 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gattinoni L, et al. A human memory T cell subset with stem cell-like properties. Nature medicine 17, 1290–1297 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lugli E, et al. Identification, isolation and in vitro expansion of human and nonhuman primate T stem cell memory cells. Nature protocols 8, 33–42 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Di Benedetto S, et al. Impact of age, sex and CMV-infection on peripheral T cell phenotypes: results from the Berlin BASE-II Study. Biogerontology 16, 631–643 (2015). [DOI] [PubMed] [Google Scholar]
  • 22.Roederer M, et al. The genetic architecture of the human immune system: a bioresource for autoimmunity and disease pathogenesis. Cell 161, 387–403 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cieri N, et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 121, 573–584 (2013). [DOI] [PubMed] [Google Scholar]
  • 24.Lugli E, et al. Superior T memory stem cell persistence supports long-lived T cell memory. The Journal of clinical investigation (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Simons BD & Clevers H Strategies for homeostatic stem cell self-renewal in adult tissues. Cell 145, 851–862 (2011). [DOI] [PubMed] [Google Scholar]
  • 26.Wherry EJ, et al. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nature immunology 4, 225–234 (2003). [DOI] [PubMed] [Google Scholar]
  • 27.Graef P, et al. Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8(+) central memory T cells. Immunity 41, 116–126 (2014). [DOI] [PubMed] [Google Scholar]
  • 28.Gattinoni L Memory T cells officially join the stem cell club. Immunity 41, 7–9 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gerlach C, et al. Heterogeneous differentiation patterns of individual CD8+ T cells. Science 340, 635–639 (2013). [DOI] [PubMed] [Google Scholar]
  • 30.Buchholz VR, et al. Disparate individual fates compose robust CD8+ T cell immunity. Science 340, 630–635 (2013). [DOI] [PubMed] [Google Scholar]
  • 31.Lanzavecchia A & Sallusto F Progressive differentiation and selection of the fittest in the immune response. Nature reviews. Immunology 2, 982–987 (2002). [DOI] [PubMed] [Google Scholar]
  • 32.Joshi NS, et al. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Cieri N, et al. Generation of human memory stem T cells after haploidentical T-replete hematopoietic stem cell transplantation. Blood 125, 2865–2874 (2015). [DOI] [PubMed] [Google Scholar]
  • 34.Roberto A, et al. Role of naive-derived T memory stem cells in T-cell reconstitution following allogeneic transplantation. Blood 125, 2855–2864 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Oliveira G, et al. Tracking genetically engineered lymphocytes long-term reveals the dynamics of T cell immunological memory. Science translational medicine 7, 317ra–198. (2015). [DOI] [PubMed] [Google Scholar]
  • 36.Fuertes Marraco SA, et al. Long-lasting stem cell-like memory CD8+ T cells with a naive-like profile upon yellow fever vaccination. Science translational medicine 7, 282ra–248. (2015). [DOI] [PubMed] [Google Scholar]
  • 37.Fuertes Marraco SA, Soneson C, Delorenzi M & Speiser DE Genome-wide RNA profiling of long-lasting stem cell-like memory CD8 T cells induced by Yellow Fever vaccination in humans. Genom Data 5, 297–301 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Biasco L, et al. In vivo tracking of T cells in humans unveils decade-long survival and activity of genetically modified T memory stem cells. Science translational medicine 7, 273ra–213. (2015). [DOI] [PubMed] [Google Scholar]
  • 39.Xu Y, et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 123, 3750–3759 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Vigano S, et al. Prolonged Antiretroviral Therapy Preserves HIV-1-Specific CD8 T Cells with Stem Cell-Like Properties. J Virol 89, 7829–7840 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Axelsson-Robertson R, Ju JH, Kim HY, Zumla A & Maeurer M Mycobacterium tuberculosis-specific and MHC class I-restricted CD8+ T-cells exhibit a stem cell precursor-like phenotype in patients with active pulmonary tuberculosis. Int J Infect Dis 32, 13–22 (2015). [DOI] [PubMed] [Google Scholar]
  • 42.Mateus J, et al. Low frequency of circulating CD8+ T stem cell memory cells in chronic chagasic patients with severe forms of the disease. PLoS Negl Trop Dis 9, e3432 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ahmed R & Akondy RS Insights into human CD8(+) T-cell memory using the yellow fever and smallpox vaccines. Immunology and cell biology 89, 340–345 (2011). [DOI] [PubMed] [Google Scholar]
  • 44.Speiser DE, et al. T cell differentiation in chronic infection and cancer: functional adaptation or exhaustion? Nature reviews. Immunology 14, 768–774 (2014). [DOI] [PubMed] [Google Scholar]
  • 45.Utzschneider DT, et al. T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 45, 415–427 (2016). [DOI] [PubMed] [Google Scholar]
  • 46.Im SJ, et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ribeiro SP, et al. The CD8(+) memory stem T cell (T(SCM)) subset is associated with improved prognosis in chronic HIV-1 infection. J Virol 88, 13836–13844 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cartwright EK, et al. Divergent CD4+ T Memory Stem Cell Dynamics in Pathogenic and Nonpathogenic Simian Immunodeficiency Virus Infections. J Immunol (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Klatt NR, et al. Limited HIV infection of central memory and stem cell memory CD4+ T cells is associated with lack of progression in viremic individuals. PLoS pathogens 10, e1004345 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cartwright EK, et al. Initiation of antiretroviral therapy restores CD4+ TSCM homeostasis in SIV-infected macaques. J Virol (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Calascibetta F, et al. Antiretroviral therapy in SIV-infected sooty mangabeys: implications for AIDS pathogenesis. J Virol (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Speiser DE, et al. Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909. The Journal of clinical investigation 115, 739–746 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Melero I, et al. Therapeutic vaccines for cancer: an overview of clinical trials. Nature reviews. Clinical oncology 11, 509–524 (2014). [DOI] [PubMed] [Google Scholar]
  • 54.De Gregorio E & Rappuoli R Vaccines for the future: learning from human immunology. Microb Biotechnol 5, 149–155 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chang JT, Wherry EJ & Goldrath AW Molecular regulation of effector and memory T cell differentiation. Nature immunology 15, 1104–1115 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pham NL, Badovinac VP & Harty JT A default pathway of memory CD8 T cell differentiation after dendritic cell immunization is deflected by encounter with inflammatory cytokines during antigen-driven proliferation. J Immunol 183, 2337–2348 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Park CO & Kupper TS The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nature medicine 21, 688–697 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mackay LK, et al. T-box Transcription Factors Combine with the Cytokines TGF-beta and IL-15 to Control Tissue-Resident Memory T Cell Fate. Immunity 43, 1101–1111 (2015). [DOI] [PubMed] [Google Scholar]
  • 59.Zhang N & Bevan MJ Transforming growth factor-beta signaling controls the formation and maintenance of gut-resident memory T cells by regulating migration and retention. Immunity 39, 687–696 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jameson SC & Masopust D Diversity in T cell memory: an embarrassment of riches. Immunity 31, 859–871 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Farber DL, Yudanin NA & Restifo NP Human memory T cells: generation, compartmentalization and homeostasis. Nature reviews. Immunology 14, 24–35 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gattinoni L The dark side of T memory stem cells. Blood 125, 3519–3520 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hosokawa K, et al. Memory Stem T Cells in Autoimmune Disease: High Frequency of Circulating CD8+ Memory Stem Cells in Acquired Aplastic Anemia. J Immunol 196, 1568–1578 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Monti P, Heninger AK & Bonifacio E Differentiation, expansion, and homeostasis of autoreactive T cells in type 1 diabetes mellitus. Curr Diab Rep 9, 113–118 (2009). [DOI] [PubMed] [Google Scholar]
  • 65.Tabler CO, et al. CD4+ memory stem cells are infected by HIV-1 in a manner regulated in part by SAMHD1 expression. J Virol 88, 4976–4986 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Buzon MJ, et al. HIV-1 persistence in CD4(+) T cells with stem cell-like properties. Nature medicine 20, 139–142 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Jaafoura S, et al. Progressive contraction of the latent HIV reservoir around a core of less-differentiated CD4(+) memory T Cells. Nature communications 5, 5407 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Nagai Y, et al. T memory stem cells are the hierarchical apex of adult T-cell leukemia. Blood (2015). [DOI] [PubMed] [Google Scholar]
  • 69.Rosenberg SA & Restifo NP Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.June CH, Riddell SR & Schumacher TN Adoptive cellular therapy: a race to the finish line. Science translational medicine 7, 280ps–287. (2015). [DOI] [PubMed] [Google Scholar]
  • 71.Dudley ME, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298, 850–854 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Brentjens RJ, et al. CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia. Science translational medicine 5, 177ra–138. (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lee DW, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385, 517–528 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kochenderfer JN, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 33, 540–549 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Brudno JN, et al. Allogeneic T Cells That Express an Anti-CD19 Chimeric Antigen Receptor Induce Remissions of B-Cell Malignancies That Progress After Allogeneic Hematopoietic Stem-Cell Transplantation Without Causing Graft-Versus-Host Disease. Journal of clinical oncology : official journal of the American Society of Clinical Oncology (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Maude SL, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England journal of medicine 371, 1507–1517 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Porter DL, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Science translational medicine 7, 303ra–139. (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Robbins PF, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clinical cancer research : an official journal of the American Association for Cancer Research 21, 1019–1027 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Robbins PF, et al. Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J Immunol 173, 7125–7130 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Pule MA, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nature medicine 14, 1264–1270 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rosenberg SA, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clinical cancer research : an official journal of the American Association for Cancer Research 17, 4550–4557 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kalos M, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science translational medicine 3, 95ra–73. (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Stevanovic S, et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 33, 1543–1550 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Turtle CJ, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Science translational medicine 8, 355ra–116. (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Busch DH, Frassle SP, Sommermeyer D, Buchholz VR & Riddell SR Role of memory T cell subsets for adoptive immunotherapy. Seminars in immunology 28, 28–34 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhou J, et al. Telomere length of transferred lymphocytes correlates with in vivo persistence and tumor regression in melanoma patients receiving cell transfer therapy. J Immunol 175, 7046–7052 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Huang J, et al. Modulation by IL-2 of CD70 and CD27 expression on CD8+ T cells: importance for the therapeutic effectiveness of cell transfer immunotherapy. J Immunol 176, 7726–7735 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Louis CU, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118, 6050–6056 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gattinoni L, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. The Journal of clinical investigation 115, 1616–1626 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Klebanoff CA, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proceedings of the National Academy of Sciences of the United States of America 102, 9571–9576 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Hinrichs CS, et al. Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proceedings of the National Academy of Sciences of the United States of America 106, 17469–17474 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wang X, et al. Comparison of naive and central memory derived CD8 effector cell engraftment fitness and function following adoptive transfer. Oncoimmunology 5, e1072671 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sommermeyer D, et al. Chimeric antigen receptor-modified T cells derived from defined CD8 and CD4 subsets confer superior antitumor reactivity in vivo. Leukemia (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Klebanoff CA, et al. Determinants of successful CD8+ T-cell adoptive immunotherapy for large established tumors in mice. Clinical cancer research : an official journal of the American Association for Cancer Research 17, 5343–5352 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Berger C, et al. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. The Journal of clinical investigation 118, 294–305 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Baitsch L, et al. Exhaustion of tumor-specific CD8(+) T cells in metastases from melanoma patients. The Journal of clinical investigation 121, 2350–2360 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Gros A, et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. The Journal of clinical investigation (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lugli E, et al. Subject classification obtained by cluster analysis and principal component analysis applied to flow cytometric data. Cytometry A 71, 334–344 (2007). [DOI] [PubMed] [Google Scholar]
  • 99.Appay V, et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nature medicine 8, 379–385 (2002). [DOI] [PubMed] [Google Scholar]
  • 100.Mackall CL, et al. Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy. Blood 89, 3700–3707 (1997). [PubMed] [Google Scholar]
  • 101.Singh N, Perazzelli J, Grupp SA & Barrett DM Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies. Science translational medicine 8, 320ra–323. (2016). [DOI] [PubMed] [Google Scholar]
  • 102.Turtle CJ, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. The Journal of clinical investigation (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang X, et al. Phase I studies of central-memory-derived CD19 CAR T cell therapy following autologous HSCT in patients with B-cell NHL. Blood (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Li Q, et al. A central role for mTOR kinase in homeostatic proliferation induced CD8+ T cell memory and tumor immunity. Immunity 34, 541–553 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hinrichs CS, et al. IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy. Blood 111, 5326–5333 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.van der Waart AB, et al. Inhibition of Akt signaling promotes the generation of superior tumor-reactive T cells for adoptive immunotherapy. Blood 124, 3490–3500 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Crompton JG, et al. Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. Cancer research 75, 296–305 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Gattinoni L, Klebanoff CA & Restifo NP Pharmacologic induction of CD8+ T cell memory: better living through chemistry. Science translational medicine 1, 11ps–12. (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Davila ML, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Science translational medicine 6, 224ra–225. (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Grupp SA, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. The New England journal of medicine 368, 1509–1518 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sabatino M, et al. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Klebanoff CA, et al. Memory T cell-driven differentiation of naive cells impairs adoptive immunotherapy. The Journal of clinical investigation 126, 318–334 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Klebanoff CA, Gattinoni L & Restifo NP Sorting through subsets: which T-cell populations mediate highly effective adoptive immunotherapy? J Immunother 35, 651–660 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ding ZC, et al. IL-7 signaling imparts polyfunctionality and stemness potential to CD4(+) T cells. Oncoimmunology 5, e1171445 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Provasi E, et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nature medicine 18, 807–815 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Stemberger C, et al. Lowest numbers of primary CD8(+) T cells can reconstitute protective immunity upon adoptive immunotherapy. Blood 124, 628–637 (2014). [DOI] [PubMed] [Google Scholar]
  • 117.Schmueck-Henneresse M, et al. Peripheral blood-derived virus-specific memory stem T cells mature to functional effector memory subsets with self-renewal potency. J Immunol 194, 5559–5567 (2015). [DOI] [PubMed] [Google Scholar]
  • 118.Terakura S, et al. Generation of CD19-chimeric antigen receptor modified CD8+ T cells derived from virus-specific central memory T cells. Blood 119, 72–82 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Becker TC, et al. Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. The Journal of experimental medicine 195, 1541–1548 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Schluns KS, Williams K, Ma A, Zheng XX & Lefrancois L Cutting edge: requirement for IL-15 in the generation of primary and memory antigen-specific CD8 T cells. J Immunol 168, 4827–4831 (2002). [DOI] [PubMed] [Google Scholar]
  • 121.Yi JS, Du M & Zajac AJ A vital role for interleukin-21 in the control of a chronic viral infection. Science 324, 1572–1576 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Frohlich A, et al. IL-21R on T cells is critical for sustained functionality and control of chronic viral infection. Science 324, 1576–1580 (2009). [DOI] [PubMed] [Google Scholar]
  • 123.Yi JS, Ingram JT & Zajac AJ IL-21 deficiency influences CD8 T cell quality and recall responses following an acute viral infection. J Immunol 185, 4835–4845 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Cui W, Liu Y, Weinstein JS, Craft J & Kaech SM An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity 35, 792–805 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.van der Windt GJ, et al. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36, 68–78 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sukumar M, et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. The Journal of clinical investigation 123, 4479–4488 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Liu L, et al. Novel CD4-Based Bispecific Chimeric Antigen Receptor Designed for Enhanced Anti-HIV Potency and Absence of HIV Entry Receptor Activity. J Virol 89, 6685–6694 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Ali A, et al. HIV-1-Specific Chimeric Antigen Receptors Based on Broadly-Neutralizing Antibodies. J Virol (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Gattinoni L & Restifo NP Moving T memory stem cells to the clinic. Blood 121, 567–568 (2013). [DOI] [PubMed] [Google Scholar]
  • 130.Kahn M Can we safely target the WNT pathway? Nature reviews. Drug discovery 13, 513–532 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ramishetti S, et al. Systemic Gene Silencing in Primary T Lymphocytes Using Targeted Lipid Nanoparticles. ACS nano 9, 6706–6716 (2015). [DOI] [PubMed] [Google Scholar]
  • 132.Zhou J, et al. Selection, characterization and application of new RNA HIV gp 120 aptamers for facile delivery of Dicer substrate siRNAs into HIV infected cells. Nucleic acids research 37, 3094–3109 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Tebas P, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med 370, 901–910 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Dragic T, et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5 [see comments]. Nature 381, 667–673 (1996). [DOI] [PubMed] [Google Scholar]
  • 135.Scholz G, et al. Modulation of mTOR Signalling Triggers the Formation of Stem Cell-like Memory T Cells. EBioMedicine 4, 50–61 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Crompton JG, et al. Lineage relationship of CD8(+) T cell subsets is revealed by progressive changes in the epigenetic landscape. Cell Mol Immunol 13, 502–513 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Chang JT, et al. Asymmetric proteasome segregation as a mechanism for unequal partitioning of the transcription factor T-bet during T lymphocyte division. Immunity 34, 492–504 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Lin WH, et al. Asymmetric PI3K Signaling Driving Developmental and Regenerative Cell Fate Bifurcation. Cell reports 13, 2203–2218 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Verbist KC, et al. Metabolic maintenance of cell asymmetry following division in activated T lymphocytes. Nature 532, 389–393 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Pollizzi KN, et al. Asymmetric inheritance of mTORC1 kinase activity during division dictates CD8(+) T cell differentiation. Nature immunology 17, 704–711 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Jeannet G, et al. Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8 T cell memory. Proceedings of the National Academy of Sciences of the United States of America 107, 9777–9782 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Zhou X, et al. Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity 33, 229–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zhao DM, et al. Constitutive activation of Wnt signaling favors generation of memory CD8 T cells. J Immunol 184, 1191–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Boudousquie C, et al. Differences in the transduction of canonical Wnt signals demarcate effector and memory CD8 T cells with distinct recall proliferation capacity. J Immunol 193, 2784–2791 (2014). [DOI] [PubMed] [Google Scholar]
  • 145.Araki K, et al. mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108–112 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Rao RR, Li Q, Odunsi K & Shrikant PA The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin. Immunity 32, 67–78 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Pearce EL, Poffenberger MC, Chang CH & Jones RG Fueling immunity: insights into metabolism and lymphocyte function. Science 342, 1242454 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.van der Windt GJ, et al. CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability. Proceedings of the National Academy of Sciences of the United States of America 110, 14336–14341 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Sukumar M, et al. Mitochondrial Membrane Potential Identifies Cells with Enhanced Stemness for Cellular Therapy. Cell metabolism 23, 63–76 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Buck MD, et al. Mitochondrial Dynamics Controls T Cell Fate through Metabolic Programming. Cell 166, 63–76 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Morrison SJ & Spradling AC Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell 132, 598–611 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Di Rosa F Two Niches in the Bone Marrow: A Hypothesis on Life-long T Cell Memory. Trends in immunology (2016). [DOI] [PubMed] [Google Scholar]
  • 153.Alp OS, et al. Memory CD8 T cells colocalize with IL-7 stromal cells in bone marrow and rest in terms of proliferation and transcription. European journal of immunology (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Becker TC, Coley SM, Wherry EJ & Ahmed R Bone marrow is a preferred site for homeostatic proliferation of memory CD8 T cells. J Immunol 174, 1269–1273 (2005). [DOI] [PubMed] [Google Scholar]
  • 155.Mazo IB, et al. Bone marrow is a major reservoir and site of recruitment for central memory CD8+ T cells. Immunity 22, 259–270 (2005). [DOI] [PubMed] [Google Scholar]
  • 156.Takada K & Jameson SC Naive T cell homeostasis: from awareness of space to a sense of place. Nature reviews. Immunology 9, 823–832 (2009). [DOI] [PubMed] [Google Scholar]

RESOURCES