Review
Memory CD8+ T cell responses to cancer

https://doi.org/10.1016/j.smim.2020.101435Get rights and content

Highlights

  • Memory CD8+ T cell subsets mediate long-lived anti-tumor immunity.

  • Stem-like memory T cells infiltrate tumors and are capable of self-renewal.

  • TRM cells are comprised of transcriptionally functional and exhausted subsets.

  • Stem-like memory T cells and TRM cells in tumors portend improved patient prognosis.

Abstract

Long-lived memory CD8+ T cells play important roles in tumor immunity. Studies over the past two decades have identified four subsets of memory CD8+ T cells - central, effector, stem-like, and tissue resident memory - that either circulate through blood, lymphoid and peripheral organs, or reside in tissues where cancers develop. In this article, we will review studies from both pre-clinical mouse models and human patients to summarize the phenotype, distribution and unique features of each memory subset, and highlight specific roles of each subset in anti-tumor immunity. Moreover, we will discuss how stem-cell like and resident memory CD8+ T cell subsets relate to exhausted tumor-infiltrating lymphocytes (TIL) populations. These studies reveal how memory CD8+ T cell subsets together orchestrate durable immunity to cancer.

Introduction

The use of CD8+ T cells has been a focus of clinical cancer therapy for over 20 years [1]. Based on their ability to clonally expand and exert cytotoxic function, T effector (TEFF) cells have long been recognized as important mediators of tumor protection [2,3]. However, studies in mice and humans have also revealed that the presence of tumor infiltrating TEFF cells is not sufficient for tumor rejection because of apoptosis induced clonal contraction [[4], [5], [6]]. Cancer is a chronic disease and, unlike immunity to acute infections, curative immunity to cancer is thought to require long-lived T cell immunity. Indeed, the persistence of adoptively transferred T cells in peripheral blood has been shown to correlate with tumor regression in cancer patients [7]. In contrast to TEFF cells, memory CD8+ T cells are capable of durably persisting and functioning throughout host tissues and tumors, making them a gold standard for anti-tumor immunity.

The earliest evidence for the generation of protective immune memory to cancer comes from pre-clinical studies showing that mice cured from primary tumors could maintain protection against secondary tumor rechallenge [[8], [9], [10]]. This persistent tumor immunity was mediated by CD8+ memory T cells, supported by the finding that depletion of CD8+ T cells eliminated tumor protection [8,11]. Early work involving peptide vaccination in humans separately showed that melanoma antigen specific CD8+ T cells could expand dramatically, acquire a CD27 CD28- CD57+ CD45RAlo memory phenotype, and demonstrate robust recall function in vitro [12]. An additional study examining melanoma patients after peptide vaccination revealed Melan-A/MART-1 specific memory T-cell responses capable of producing IL-2, IFN-γ and GM-CSF [13]. These early clinical and preclinical studies collectively revealed the possibility of generating functional T-cell memory against cancer, and the importance of such responses in curative immunity. However, the dynamic and heterogeneous nature of memory CD8+ T cell responses, throughout host lymphoid and peripheral tissue compartments, was only beginning to be explored.

Priming of naïve CD8+ T cells induces clonal expansion and the acquisition of peripheral tissue homing capacity, effector function and cytolytic activity. After contraction of the TEFF response, memory T cells continue to recirculate but also become durably resident in tissues throughout the body. Memory CD8+ T cells persist as a heterogenous population including circulating memory (TCIRC) subsets- central memory (TCM), effector memory (TEM) and stem cell memory T cells (TSCM), and a non-circulating subset known as resident memory T cells (TRM) (Fig. 1). TCM and TSCM cells express molecules such as CD62L and CCR7 that enable their access to bone marrow (BM) and secondary lymphoid organs (SLO), whereas TEM cells lack lymphoid tissue homing receptors, and preferentially recirculate through blood and peripheral tissues [[14], [15], [16]]. TRM cells lack markers of circulating subsets, instead expressing tissue adhesion markers such as CD103 and CD69 [17].

The maintenance and differentiation of these memory subsets are mediated by both cell extrinsic and intrinsic factors. Cytokines such as IL-2 promote cell proliferation during priming, and is essential for functional CD8+ memory T cell recall responses [18], whereas IL-7 and IL-15 promote survival, differentiation and maintenance of memory [19]. Expression of various transcription factors also controls the differentiation and function of distinct memory CD8+ T cell subsets. For instance, KLF2 promotes the expression of CD62L and S1PR1 which are essential for TCIRC tissue egress and homing to SLOs [20]. In contrast, Hobit, Blimp1 [21,22] and Runx3 [23] govern TRM cell differentiation by limiting tissue egress. Regardless, memory T cell subsets are capable of differentiating and interconverting in vivo (Fig. 1). Whereas TSCM cells are considered the most pluripotent, TCM cells are capable of giving rise to TEM and TRM subsets [[24], [25], [26]]. Moreover, recent data suggest that TRM cells can become TCM and TEM cells following local reactivation [27]. Collectively, extrinsic and intrinsic factors within a given microenvironment regulates the distinct roles and differentiation pathways for memory T-cell subsets.

The growing knowledge of these subsets from studies in infectious disease models continues to inform the field of cancer immunology. Indeed, each of these subsets plays a unique role in immunity to cancer, and recent studies have revealed the heterogeneity that governs their differentiation and maintenance. The goal of this review article is to summarize the characteristics and anti-tumor function of different subsets of memory CD8+ T cells. We will discuss how these populations can coordinate to provide long-term immunity to cancer while underscoring the association between the presence of memory CD8+ T cells and improved outcomes for cancer patients.

As described above, two major circulating memory CD8+ T cell subsets are TCM and TEM cells. First described in humans in 1999, these subsets were initially differentiated by their expression of CD62L [28] and CCR7 [29] which are essential for T cell migration to lymph nodes. However, these populations are now well distinguished by their unique phenotypes, functions, and migratory properties. TCM cells exhibit a CD45RA CD45RO+ CCR7+ CD62L+ phenotype in humans, and are defined by their lymph node homing properties [30,31]. They exhibit a higher proliferative capacity than their TEM counterparts which exhibit a CD45RA CD45RO+ CCR7 CD62L phenotype and, accordingly, are excluded from lymph nodes, while expressing integrins and chemokine receptors necessary for localization to inflamed tissues [30,32]. In mice, the phenotype and localization of TCM and TEM subsets largely overlap with those in humans, with the exception that CD44+ is used as a memory marker instead of CD45RA/CD45RO [33]. TCM and TEM cells each respond to TCR stimulation in a different manner. Upon anti-CD3/CD28 stimulation in vitro, TEM cells secrete large amounts of effector molecules including IFN-γ and perforin, whereas TCM cells predominantly express high levels of IL-2, clonally expand, and are capable of differentiating into both TEFF and TEM cells [30,[34], [35], [36]]. Thus, TCM and TEM each play an important and distinctive role in the memory response.

Memory T cell fate is determined by distinct transcriptional regulators. KLF2 has been shown to directly activate the promoters of S1P1 and CD62L, which are important for tissue egress and homing to lymph nodes [20]. T-box transcription factors T-bet and its homolog Eomes are master regulators for the differentiation of memory CD8+ T cells [37]. While low levels of T-bet are required for the development of KLRG1 IL-7R+ memory precursor cells, high levels of T-bet have been shown to suppress the expression of IL-7R, which preferentially drives the differentiation of TEM cells [38,39]. On the other hand, Eomes has been found to be essential for TCM differentiation, as well as maintaining the longevity and rapid recall responses of TCM cells [40]. The transcription factors Blimp-1 and Id2 induce terminal differentiation thus promoting CD8+ T cells differentiation into TEM cells, whereas Id3 correlates with the upregulation of Il-7r, Sell, Il2, Bcl2, and Ccr7, genes essential for the differentiation of CD8+ T cells into a TCM phenotype [[41], [42], [43]]. Thus, TCM and TEM cells represent transcriptionally distinct lineages.

TCM cells have been shown to have more potent anti-tumor activity than TEFF cells. Using the transplantable B16 murine melanoma model, the adoptive transfer of IL-15 cultured transgenic pmel CD8+ TCM cells was shown to be significantly more effective in controlling established tumors, as compared with IL-2 cultured TEFF/TEM-like cells [44]. Transferred TCM pmel cells also gave rise to 14-fold larger populations in blood and spleen [44]. In a similar preclinical model, the adoptive transfer of IL-21 cultured pmel cells, which also had a TCM phenotype, displayed enhanced antitumor responses compared to those cultured with IL-2 [45]. Similar observations were found using CMV-pp65 specific T cells taken from human peripheral blood, with TCM cells providing more robust antitumor immunity than TEM cells upon transfer into immunodeficient mice bearing CMV-pp65-expressing leukemia [46]. These data establish the superior anti-tumor function of TCM cells compared to TEM and TEFF cells, consistent with their improved capacity to engraft and clonally expand [44,46].

Despite the potency of TCM cells on a per-cell basis, TEM cells also represent a vital component of the anti-tumor response, particularly when generated in high frequencies. In vitro-generated pmel TEM cells clonally expand, secrete high levels of IFN-γ, and are capable of regressing large, established B16 tumors [47]. Several studies have shown that in vivo vaccination can promote tumor-Ag specific TEM cells which sustain the ability to secret IFN-γ and induce tumor regression [[48], [49], [50], [51]]. Interestingly, a persisting mouse gamma herpesvirus type 68 infection, which preferentially induces a TEM response [50,52], was shown to be a more effective tumor vaccine than an acute gamma herpesvirus which induces a TCM response [50]. Similarly a cytomegalovirus-based cancer vaccine, which is known to generate large frequencies of TEM-like inflationary memory CD8+ T cells [53], was shown to be more potent against cancer as compared to a vesicular somatitis viral vaccine that does not induce inflationary memory [54]. In the context of B16 melanoma-bearing mice that develop autoimmune vitiligo, large populations of melanoma-specific TEM cells persist in the spleen, in conjunction with long-lived protective immunity against melanoma in the lungs [55]. Such studies collectively show that tumor-specific TEM responses can be generated in large frequencies, and can remain functional against cancer, even in settings of persistent antigen exposure.

The presence of TCM and TEM cells has been associated with better prognosis in cancer patients. Indeed anti-CTLA-4 immunotherapy has been shown to augment CD8+ memory T cell formation and maintenance in mouse models of infection, and may thus also promote memory to cancer [56]. In advanced melanoma patients on immune checkpoint blockade (ICB) therapy with anti-CTLA4, a higher proportion of CD45RO+ memory cells out of total CD8+ T cells in blood was a positive predictor for both response rate and overall survival [57]. PD-1 is more highly expressed on TEM cells than TCM cells [58], although both populations were able to predict patient responses to anti-PD1 immunotherapy [59,60]. In advanced melanoma and non-small cell lung cancer (NSCLC) patients, the frequency of CD45RA CCR7+ TCM cells in blood was shown to be a positive predictor for both response to anti-PD1 therapy and survival [59]. Responders that had a higher TCM/TEFF ratio in blood also had a higher inflammatory gene expression profile in their tumors [59], suggesting the importance of TCM cells in anti-tumor immune responses in patients. In other studies, TEM cells were specifically associated with better responses to ICB therapy and improved survival. In advanced melanoma patients, the number of highly expanded CD8+ T cell clones (clonal frequency>0.5 %) in blood 21 days after the initiation of ICB treatment was found to be associated with better clinical response; these clones mainly possessed a CD45RA CD27 TEM phenotype [60]. A detailed study of the TEM population in advanced melanoma patients after anti-CTLA-4 found that, instead of a more terminally differentiated CD45RO+ CD62L CD27 CD28 TEM population, it was the less differentiated CD45RO+ CD62L CD27+ CD28+ TEM subset that was associated with both better clinical response and overall survival [61]. Thus, the above studies show that in addition to TCM, TEM cells are also likely to play a key role in anti-tumor immunity.

TCM cells were considered the ‘stem-like’ memory population until a less differentiated memory stem cell- TSCM population was identified in 2005. TSCM cells were first described in a mouse graft versus host disease (GVHD) model as a CD44 CD62L+ naïve-like population, even though antigen experienced, and expressed high levels of stem cell antigen 1 (Sca-1), IL-2Rβ/IL-15Rα, and the anti-apoptotic transcription factor Bcl-2 [62]. These naïve-like memory cells exhibited a greater ability to self-proliferate, differentiated into TEM, TCM and TEFF cells, and induced stronger GVH responses in mice, compared to TCM and TEM cells [62]. The presence of long-lived TSCM cells was subsequently identified in humans within the naïve-like CD8+ T-cell compartment [63]. These cells maintained a CD45RA+ CD45RO CCR7+ CD62L+ CD27+ CD28+ IL-7Rα+ naïve-like phenotype, while also expressing memory markers CD95 and IL-2Rβ [63]. Additional transcriptomic analyses revealed that these human TSCM cells had a transcriptional profile more closely related to human TCM than to naïve T cells, with similar levels of TCR rearrangement excision circles (TRECs) compared to TCM cells, indicating that TSCM cells are not recent thymic emigrants [63]. TSCM cells were subsequently identified in non-human primates where they served as precursors for TCM cells [64]. Additionally, TSCM cells were shown to exhibit enhanced proliferation when re-exposed to antigen while continuing to self-renew as TSCM, demonstrating a true stemness [65] (Fig. 1). TSCM cells are extremely durable as evidenced by a yellow fever vaccine study wherein induced TSCM cells were shown to persist for decades [66]. These features of self-renewal, persistence and multipotency could lead to the strong anti-tumor function of TSCM cells.

The anti-tumor function of TSCM was first demonstrated by studies involving in vitro cultured pmel T cells against mouse B16 melanoma. By enhancing the Wnt-β-catenin signaling pathway, the expression of Tcf1 and Lef1 was induced, thus promoting the differentiation of pmel TSCM cells with a CD62LhiCD44 phenotype [67]. Adoptive transfer of these cells resulted in greater regression of melanoma tumors compared with mice transferred with TCM or TEM cells [67].

The TSCM features of self-renewal and plasticity have led to investigations of human chimeric antigen receptor (CAR) TSCM cells as a potential therapeutic strategy for blood cancers. Human anti-CD3/anti-CD28 activated CD8+ T cells cultured with IL-7 and IL-15 and transduced with a γ retroviral vector encoding a CD19-targeting CAR resulted in a higher proportion of TSCM cells compared with IL-2 induced CAR-T cells [68]. Multiple strategies have been used to generate clinical-grade CAR TSCM cells. Activation of naïve T cells using the glycogen synthase-3β (GSK-3β) inhibitor TWS119 in the presence of IL-7 and IL-21 prior to transduction with a CD19-CAR, resulted in a primarily TSCM population [69]. In another study, previously engineered CD19-CAR-T cells were expanded by TCR stimulation and then cocultured with Notch ligand Delta-like 1-expressing cells in the presence of IL-7, which resulted in conversion into TSCM cells [70]. The enhanced NOTCH signaling promoted telomere elongation, leading to enhanced in vivo persistence as compared to conventional CAR-T cells [70]. In preclinical studies, CAR TSCM mediated improved anti-tumor immunity compared to conventional CAR-T cells [[68], [69], [70]]. Multicenter, phase 1 and 2 trials are underway to test the safety and efficacy of CAR-modified TSCM cells in patient with blood malignancies.

Naïve-like TSCM cells are a circulating T-cell population and, accordingly, are excluded from peripheral tissues and solid tumors [64,71]. Despite this, several studies have demonstrated that solid tumors are occupied by populations of TCF1+ (encoded by TCF7) stem-like memory T cells with an antigen experienced phenotype [[71], [72], [73]]. Mouse and human tumors were found to contain TCF1+PD-1+ GzmB CD8+ T cells [72]. In a mouse B16-gp33 melanoma model, TCF1+ TILs expressed memory markers including CXCR5, Ly108(Slamf6), and CD62L, as well as PD-1, CTLA-4, and Lag3 [72]. The expression of these checkpoint molecules on stem-like TILs suggest that this subset could be an important target for anti-PD1 and anti-CTLA4 treatment. Indeed, after adoptive transfer, the gp33-specific TCF1+ TILs were able to expand and differentiate into TCF1 cells while maintaining secretion of IL-2 and TNF-α, and the durable anti-tumor effects of anti-PD1 and anti-CTLA4 therapy in mice were found to be dependent on Tcf7 expression in T cells [72].

High-dimensional flow cytometry also confirmed stem-like CD8+ TILs in lung cancer patients. Human stem-like CD8+ TILs upregulated TCF7 CXCR5 EOMES BCL6, FOXO1 and CD28, and also expressed PDCD1 and TIGIT. The CXCR5+ TILs rapidly produced IFN-γ, IL-2 and TNF-α upon PMA/ionomycin stimulation and extensively proliferated and differentiated into a CXCR5 population. Furthermore, the abundance of CXCR5+ CD8+ T cells in tumor correlated with better disease control [71]. Concordantly, in melanoma patients, whereas TCF1+ CD8+ TILs express lower levels of PD-1 than their TCF1 TIL counterparts [63], the presence of TCF1+ CD8+ TILs is associated with better clinical response to ICB therapy [74]. The above studies highlight the role of stem-like TILs in tumor control and response to immunotherapy.

The features of tumor microenvironments that support stem-like T cells are just beginning to be understood. Interestingly, an abundance of potassium in the tumor microenvironment (TME) has recently been implicated as an important factor. Tumor necrosis, which has been shown to be inversely correlated with patient survival [75,76], leads to increased levels of extracellular potassium in the TME which suppresses T cell effector function [77]. However, treatment of pmel CD8+ TILs with elevated potassium concentrations resulted in differentiation of TCF1+ T cells, which resulted in regression of established tumors [77]. This study further shows the importance of stem-like TILs in an immune-suppressive TME.

Stem-like CD8 + TILs are phenotypically similar to tumor-infiltrating populations that have been termed progenitor exhausted (Texprog) cells [78], and these populations are likely to be overlapping [79]. Texprog is a heterogenous population as revealed by a recent study in a LCMV mouse model and human melanoma tumors [78]. Four developmentally related subsets were identified, of which two subsets were the less differentiated Texprog cells that expressed high or intermediate levels of TCF1. Functionally, these subsets had the ability to interconvert, and both populations could give rise to Texprog, as well as terminally and intermediate exhausted subsets [78]. In human kidney tumors, TCF1+ stem-like TILs had a high degree of TCR overlap with TCF terminally differentiated populations, thus supporting the idea of stem-like T cell precursors [80]. In another study, a similar Tcf7, Cxcr5, Ccr7, Sell, Il7r, Cd28 expressing Texprog population was found in the B16-OVA model [73]. Upon treatment with anti-PD-1, these Texprog cells expanded to a larger extent, and expressed higher levels of TNF-α and IL-2, compared to terminally exhausted cells. This study also showed that Texprog cells in the spleen and lymph nodes could persist in the absence of tumor antigen, indicating that they are true memory T cells. As with prior studies focused on TCF1+ stem-like cells [72], anti-PD-1 treatment expanded the number of Texprog cells in tumors [73]. Interestingly, in human tumors, TCF1+ CD8+ TILs preferentially reside in regions of APC dense zones which resemble T-cell zones of lymphatic tissue, or intra-tumoral immune niches [80]. In patients who had disease progression, there were >10-fold fewer intra-tumoral immune niches [80]. Thus, the critical importance of Texprog cells for anti-tumor immunity has emerged. However, the durability of these stem-like cells in tumors of cancer patients remains to be elucidated.

Pioneering work in the early 2000s discovered memory CD8+ T cells in peripheral tissues, which were initially thought to be TEM in recirculation from the blood [81,82]. Early findings in models of vesicular stomatitis virus and listeria monocytogenes infections [82] revealed that these T cells were actually durably resident in tissues, and such resident memory (TRM) responses have since been documented in response to multiple pathogens [[83], [84], [85], [86], [87], [88], [89], [90]]. TRM cells lack expression of CD62L (L-selectin); which differentiates them from naïve T cells and TCM cells that require CD62L for entry into secondary lymphoid organs [31]. In order to differentiate TRM cells from effector and TEM cells, tissue retention markers, CD49a (VLA-1) and CD103, are typically used. CD49a promotes tissue retention and survival through binding to collagenase type IV [91], while CD103 is a TGF-ß induced molecule that promotes TRM cell tissue retention by binding to E-cadherin [92]. TRM cells are also known to express CD69, a marker of T cell activation, which blocks T cell expression of S1PR1 [83], and lack the expression of CCR7, which cooperates with S1PR1 for tissue egress, thereby promoting tissue retention and residency [93,94]. While CD49a, CD103, and CD69 are typical markers of tissue residency, mouse studies have shown that they are not absolute nor required [88,[95], [96], [97], [98]]. However, CD69 appears to be a reliable TRM marker in humans, with tissue TRM cells in healthy individuals overwhelmingly expressing CD69, but not CD103 [21].

A unique transcriptional program regulating TRM development has also been well established [23,83,99]. As previously described, Hobit, Blimp1 [22] and Runx3 [23] were found essential for the development of TRM cells in mice, while in humans, Hobit was dispensable [21]. Loss of KLF2, which regulates S1PR1, has been shown to enhance tissue residency [100]. Taken together, these findings have revealed TRM cells as a distinct memory lineage.

TRM cells comprise the largest memory compartment in human adults and have been identified in almost all epithelial and mucosal tissues where cancers arise [101,102]. In some cases, TRM cells accumulate at sites of antigen persistence [103] and adapt to their surroundings where they reside. In anatomical regions with a high cell turnover rate, such as the lamina propria of the gut, immune cells such as macrophages support the formation of TRM aggregates [104]. In other barrier tissues, TRM cells occupy de novo niches, such as repair-associated memory depots in the lung, and mucosa-associated lymphoid tissue in the female reproductive tract [105,106]. Localization to barrier sites of mucosal tissues exerts a metabolic burden that typically limits the persistence of T cells. However, TRM cells utilize fatty acid beta-oxidative phosphorylation to support their longevity [[107], [108], [109]]. In contrast to conventional memory T cells which conduct their own fatty acid synthesis, TRM cells in the skin have been shown to express high levels of fatty acid binding proteins FABP4 and 5, to facilitate the necessary uptake of fatty acids [107]. These metabolic characteristics of TRM cells that support their function in peripheral tissues may also support their function in tumors. TRM cells in the tumor microenvironment must compete for nutrients with tumor cells, which use high levels of glucose and glutamine [110]. Reliance on fatty acid catabolism has recently been shown to be essential for CD8+ TIL function [111]. The PPAR agonists fenofibrate, and bezafibrate, which both promote fatty acid oxidation, have each been shown to improve T cell anti-tumor activity [111,112], although it remains to be seen if these effects are due to improved TRM responses. Regardless, the metabolic requirements of TRM cells may make them ideally suited to persist and function in a metabolically hostile TME.

A substantial body of work from murine tumor models has revealed a central role for TRM cells in immunity to cancer. A study in 2010 first introduced the concept of tissue-localized tumor immunity in response to infection through various routes with recombinant vaccinia virus expressing OVA257–264 [113]. Optimal protection against dermal B16-OVA rechallenge was only afforded by prior infection in the skin [113]. This study further identified OVA-specific T cells in the skin, and referred to them as “skin-resident TEM cells” [113].

Studies in 2017 established that tumor Ag-specific memory T cells can become durably resident in the skin, where they are necessary for long-lived protection against B16 melanoma [114]. Following Treg cell depletion and surgical excision of dermal melanoma, mice that developed autoimmune vitiligo harbored large skin populations of CD69+ CD103+ CD8+ TRM cells specific for the melanoma/melanocyte shared antigen gp100 [114]. The absence of CD103 on CD8+ T cells impaired the establishment of TRM cells and eliminated tumor protection [114]. The concept of generating skin TRM responses against tumor/self-antigens was subsequently illustrated in studies involving intradermal DNA vaccination against gp100 [115].

Additional studies solidified a role of TRM cells in immunity directed against tumors expressing viral antigens. TRM cells were implicated in a human papilloma virus E7 vaccine-induced response against orthotopic TC1 head and neck tumors [116]. Use of the drug FTY720, which blocks T cell egress from lymph nodes, in conjunction with elegant parabiosis studies, revealed a minimal role for circulating T cells in tumor protection [116]. In mice bearing B16 tumors expressing a viral antigen, TRM cells were shown to sustain tumor equilibrium and maintain microscopic tumors in the skin [117]. This was in contrast to mice lacking CD69 or CD103 in which TRM responses could not be generated, and macroscopic tumors grew, illustrating that TRM cells can also mediate cancer immune surveillance [117]. Despite this, TRM cells may not function in isolation, as a thorough investigation of VACV-OVA vaccination strategies showed that the interplay between TRM and TCIRC cells was required for optimal tumor immunity in the skin [26]. Indeed, more recent studies have shown that CD8+ TRM cells facilitate antigen shedding by directly killing tumor cells, thereby allowing dendritic cells (DCs) to relay tumor antigens to the draining lymph node to further prime CD8+ T cells [118]. Collectively, these mouse studies illustrate a crucial and multi-faceted role of TRM cells in mediating cancer immunity and immune surveillance.

TRM cells have recently been shown to exist in multiple human solid tumors. Prior to use of the term “resident memory,” CD103 expressing CD8 T cells were identified within multiple types of human tumors [[119], [120], [121], [122]]. With CD103 now recognized as a common TRM cell marker, these studies can be viewed as the earliest evidence of TILs having TRM-like properties. Additional characterization of CD8+CD103+ TILs in high grade serous ovarian cancer (HGSOC) and endometrial adenocarcinoma showed elevated expression of the exhaustion marker PD-1 [[123], [124], [125]]. CD8+ T cells in pediatric glial tumors exhibited a CD45RO+ CD69+ CCR7 TRM-like phenotype, in addition to multiple inhibitory checkpoints including PD-1, and TIGIT [126]. Using an immunofluorescence technique to visualize TRM in NSCLC, co-expression of CD49a (VLA-1) was identified on CD103+CD8+ T cells [116]. This study also showed elevated levels of PD-1 and TIM-3 on CD103+CD8+ TILs as compared to CD103 TILs. Similarly, in CD103+ CD8+ TRM cells in mesothelioma tumors showed higher expression of PD1 and TIM-3 as well as more IFN-γ production compared to CD103 TILs [127]. Accordingly Cy-TOF analysis of melanoma-infiltrating T cells showed that a CD69+ subset (among which ∼50 % expressed CD103), co-expressed high levels of inhibitory checkpoint molecules CTLA-4 and PD-1 [128]. The above phenotypic analyses demonstrated the presence of TRM-like TILS within the tumor microenvironment.

Transcriptional profiling has further supported the identification of TRM cells in tumors. NSCLC tumors with a high TIL infiltration score were shown to have more pronounced gene expression characteristics of TRM cells including higher transcript levels for ITGAE (CD103), CD69, ITGA1 (CD49a), CXCR6, PDCD1 (PD-1), HAVCR2 (TIM3), LAG3, and TIGIT, but lower expression of KLRG1, CCR7, SELL (CD62 L), and S1P1 [129]. With the growing availability of single cell RNA sequencing (scRNA-seq), populations of TRM cells have been more precisely characterized. scRNA-seq of TILs isolated from two patients with triple negative breast cancer (TNBC), coupled with bulk RNA-seq data on sorted TIL populations, showed that CD8+CD103+ TILs exhibited multiple features of TRM differentiation [130]. Another study sequenced TILs from twelve NSCLC tumors and identified clusters with high expression of the TRM specific transcription factors CD69, CD103 and CXCR6 [131], and a recent study sequenced TILS from fourteen treatment naïve patients across four different tumor types and similarly identified CD103+ ZNF683+ (Hobit) TRM cells that also had expression of exhaustion markers [132]. Interestingly, when looking at a clonally expanded PD1-expressing TRM subset in tumors from lung cancer patients, the TRM, but not non-TRM cells, were transcriptionally and epigenetically enriched for superior cytotoxicity and functionality [133], suggesting that TRM are an important component of the anti-tumor immune response.

Evidence that CD8+CD103+ T cells had prognostic value for patients did not appear in the literature until 2014, when it was shown that this subset was strongly associated with survival in HGSOC [123]. Interestingly, the presence of CD8+CD103 cells conferred no benefit when compared to tumors devoid of all CD8+ T cells, suggesting that the CD103+ CD8+ subset dominates the protective response in HGSOC [123]. Similar correlation between CD103+CD8+ T cells and improved prognosis has also been shown in urothelial cancer [134], breast cancer [135], endometrial cancer [125], and lung cancer [116,136,137]. Even when CD69+ CD103+ PD1+ LAG3+ TRM cells with high levels of immune checkpoint molecule expression were found in treatment-naïve melanoma tumors, these cells significantly expanded early during anti-PD1 immunotherapy, and the number of CD69+ CD103+ TRM cells was associated with improved patient survival [138]. However, this same association has not been true for pancreatic cancer. While CD103+ TRM TILs have been identified in pancreatic ductal cell adenocarcinoma (PDAC) [139,140], CD103+CD8+ TIL numbers did not predict survival, nor did CD103+ CD8+ TILs in intratumoral regions [139]. Interestingly though, a high ratio of CD103+ TILs in intratumoral versus stromal locations was predictive of prognosis, potentially indicating the importance of spatial relationships between TRM-like cell subsets in PDAC [139]. Moreover, highlighting the importance of spatial localization, increased densities of CD103+ CD8+ TILs in cancer islands within breast tumors was found to be more significantly associated with recurrence free survival than CD8+ TILs within stroma [141].

With increased granularity of transcriptional profiling by scRNA-seq, multiple studies have directly correlated the identification of TRM in patient tumors with improvement in survival. A TRM gene signature from TNBC patients was predictive of survival in TNBC patients from the METABRIC consortium, and could be used to distinguish breast cancer responders to ICB therapy [130]. As in TNBC, a scRNA-seq derived gene signature was used to stratify patients in the TCGA lung adenocarcinoma dataset; patients with a higher expression the CD8+ HOBIThi gene signature had a significantly greater overall survival compared to other TIL derived signatures [131]. The studies above clearly show that TILs with features of TRM portend improved patient survival across multiple tumor types, emphasizing the importance of these cells in human cancer immunity.

TRM cells have now been established as an important memory subset in the human tumor microenvironment. New evidence reveals that TRM-like cells in the tumor microenvironment are a complex population, with multiple subsets [132,140,142]. In lung cancer patients, although the TRM signature including CD69, CD103 and CXCR6 was broadly enriched in all tumor TRM cells, these cells comprised three clusters: (1) Hobit-enriched CD103hi PD1lo, (2) Granzyme K-enriched CD103lo PD1hi, and (3) CD103hi PD1hi CTLAhi LAG3hi TIGIThi, the latter of which was most consistent with exhaustion [142]. This work showed that not all TRM cells express high levels of negative checkpoint molecules. Additionally, scRNA-seq analysis of CD8+ T cells from lung, endometrial, colon, and renal cancers identified a CD103+ZNF683+(Hobit) TRM subsets that similarly contained subpopulations with both high and low PD1 expression [132]. In a mouse melanoma model, two populations of TILs with tissue-resident gene signatures were identified, in which the long-lived Id3hiBlimp1lo memory-like CD8+ population had higher CD69 expression and exhibited features of progenitor exhaustion, while the Blimp1hi Id3lo effector-like population had features of terminal exhaustion [143]. Thus, within TRM subsets in tumors, some exhibit a more stem-like progenitor exhaustion profile and others are more terminally exhausted.

One could speculate that circulating CD8+ TSCM cells, unlike TEXprog cells which reside in tumors, may continuously replenish TRM cells in tumors. Indeed, a recent study showed clonotypic expansion of effector-like T cells in tumor and blood, and surmised that circulating counterparts may replenish the existing TILs with non-exhausted cells [132]. Future studies examining the clonal relationship between different TRM populations, as well as other memory T-cell subsets, within matched tissues from the same patient will be needed to broaden our understanding TRM response to human cancers.

Section snippets

Conclusions

In summary, the field has identified four memory CD8+ T cell subsets that play crucial roles in anti-tumor immunity (Fig. 1). In the past few years, a vital role for CD8+ TRM cells in providing durable anti-tumor immunity has become more appreciated. Studies using single cell transcriptomic approaches have broadened our understanding of the heterogeneity within TILs across multiple different cancer types (Fig. 2). Prior definitions of dysfunctional T cells in tumors, solely based on their

Declaration of Competing Interest

All authors declare no competing interests.

Acknowledgments

This work was funded by the American Cancer Society (CSDG18-167-01) and the Society of Surgical Oncology Clinical Investigator Award, to CVA.; NIH R01 CA225028 and The Knights of the York Cross of Honour Philanthropic Fund to MJT; a Borroughs Welcome BDLS Training Grant to JH. The views expressed are those of the authors and not necessarily those of the NIH or the American Cancer Society.

References (144)

  • C.S. Hinrichs et al.

    IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy

    Blood

    (2008)
  • X. Wang et al.

    Engraftment of human central memory-derived effector CD8+ T cells in immunodeficient mice

    Blood

    (2011)
  • C.A. Klebanoff et al.

    Programming tumor-reactive effector memory CD8+ T cells in vitro obviates the requirement for in vivo vaccination

    Blood

    (2009)
  • J.K. Tietze et al.

    The proportion of circulating CD45RO(+)CD8(+) memory T cells is correlated with clinical response in melanoma patients treated with ipilimumab

    Eur. J. Cancer

    (2017)
  • K. Wistuba-Hamprecht et al.

    Peripheral CD8 effector-memory type 1 T-cells correlate with outcome in ipilimumab-treated stage IV melanoma patients

    Eur. J. Cancer

    (2017)
  • N. Cieri et al.

    IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors

    Blood

    (2013)
  • Y. Xu 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

    (2014)
  • M. Sabatino et al.

    Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies

    Blood

    (2016)
  • K.D. Klonowski et al.

    Dynamics of blood-borne CD8 memory T cell migration in vivo

    Immunity

    (2004)
  • D.J. Zammit et al.

    Residual antigen presentation after influenza virus infection affects CD8 T cell activation and migration

    Immunity

    (2006)
  • A.J. Bankovich et al.

    CD69 suppresses sphingosine 1-phosophate receptor-1 (S1P1) function through interaction with membrane helix 4

    J. Biol. Chem.

    (2010)
  • D. Fernandez-Ruiz et al.

    Liver-resident memory CD8(+) t cells form a front-line defense against malaria liver-stage infection

    Immunity

    (2016)
  • E.M. Steinert et al.

    Quantifying memory CD8 t cells reveals regionalization of Immunosurveillance

    Cell

    (2015)
  • M.E. Dudley et al.

    Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes

    Science

    (2002)
  • D. Valmori et al.

    Circulating Tumor-reactive CD8(+) T cells in melanoma patients contain a CD45RA(+)CCR7(-) effector subset exerting ex vivo tumor-specific cytolytic activity

    Cancer Res.

    (2002)
  • A. Prevost-Blondel et al.

    Tumor-infiltrating lymphocytes exhibiting high ex vivo cytolytic activity fail to prevent murine melanoma tumor growth in vivo

    J. Immunol.

    (1998)
  • S.A. Rosenberg et al.

    Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma

    J. Immunol.

    (2005)
  • N.S. Joshi et al.

    Effector CD8 T cell development: a balancing act between memory cell potential and terminal differentiation

    J. Immunol.

    (2008)
  • P.F. Robbins et al.

    Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy

    J. Immunol.

    (2004)
  • U. Zangemeister-Wittke et al.

    Recruitment and activation of tumor-specific immune T cells in situ. CD8+ cells predominate the secondary response in sponge matrices and exert both delayed-type hypersensitivity-like and cytotoxic T lymphocyte activity

    J. Immunol.

    (1989)
  • J.D. Ellenhorn et al.

    Mechanism of tumor rejection in anti-CD3 monoclonal antibody-treated mice

    J. Immunol.

    (1990)
  • L.X. Wang et al.

    Memory T cells originate from adoptively transferred effectors and reconstituting host cells after sequential lymphodepletion and adoptive immunotherapy

    J. Immunol.

    (2004)
  • M.J. Pittet et al.

    Expansion and functional maturation of human tumor antigen-specific CD8+ T cells after vaccination with antigenic peptide

    Clin. Cancer Res.

    (2001)
  • E. Jager et al.

    Peptide-specific CD8+ T-cell evolution in vivo: response to peptide vaccination with Melan-A/MART-1

    Int. J. Cancer

    (2002)
  • D.L. Farber et al.

    Human memory T cells: generation, compartmentalization and homeostasis

    Nat. Rev. Immunol.

    (2014)
  • M.D. Martin et al.

    Defining memory CD8 t cell

    Front. Immunol.

    (2018)
  • L. Gattinoni et al.

    T memory stem cells in health and disease

    Nat. Med.

    (2017)
  • D.M. Mitchell et al.

    Distinct roles for IL-2 and IL-15 in the differentiation and survival of CD8+ effector and memory T cells

    J. Immunol.

    (2010)
  • R. Carrio et al.

    Initial antigen encounter programs CD8+ T cells competent to develop into memory cells that are activated in an antigen-free, IL-7- and IL-15-rich environment

    J. Immunol.

    (2004)
  • A. Bai et al.

    Kruppel-like factor 2 controls T cell trafficking by activating L-selectin (CD62L) and sphingosine-1-phosphate receptor 1 transcription

    J. Immunol.

    (2007)
  • L.K. Mackay et al.

    Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes

    Science

    (2016)
  • J.J. Milner et al.

    Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours

    Nature

    (2017)
  • L. Gattinoni et al.

    Paths to stemness: building the ultimate antitumour T cell

    Nat. Rev. Cancer

    (2012)
  • J.F. Osborn et al.

    Central memory CD8+ T cells become CD69+ tissue-residents during viral skin infection independent of CD62L-mediated lymph node surveillance

    PLoS Pathog.

    (2019)
  • M. Enamorado et al.

    Enhanced anti-tumour immunity requires the interplay between resident and circulating memory CD8(+) T cells

    Nat. Commun.

    (2017)
  • R. Fonseca et al.

    Developmental plasticity allows outside-in immune responses by resident memory T cells

    Nat. Immunol.

    (2020)
  • F. Sallusto et al.

    Two subsets of memory T lymphocytes with distinct homing potentials and effector functions

    Nature

    (1999)
  • F. Sallusto et al.

    Central memory and effector memory T cell subsets: function, generation, and maintenance

    Annu. Rev. Immunol.

    (2004)
  • A.D. Roberts et al.

    Differential contributions of central and effector memory T cells to recall responses

    J. Exp. Med.

    (2005)
  • L. Tussey et al.

    Functionally distinct CD8+ memory T cell subsets in persistent EBV infection are differentiated by migratory receptor expression

    Eur. J. Immunol.

    (2000)
  • Cited by (84)

    • O-GlcNAcylation at the center of antitumor immunity

      2023, Current Opinion in Biotechnology
    View all citing articles on Scopus
    View full text