Abstract
Chronic antigen stimulation leads to T cell exhaustion, and nutrient restrictions and other suppressive factors in the tumor microenvironment further exacerbate T cell dysfunction. Better understanding of heterogeneity and dynamics of exhausted CD8 T cells will guide novel therapies that modulate T cell differentiation to achieve more effective anti-tumor responses.
One-sentence summary:
Understanding the heterogeneity of exhausted CD8 T cells will help inform new therapies.
Defining T cell exhaustion
T cell exhaustion, a dysfunctional and hyporesponsive cellular state commonly observed in response to persistent antigen exposure, is a shared feature in chronic infection, cancer, and autoimmune disease (1). Originally perceived as an undesirable outcome, it has become increasingly apparent that the molecular and phenotypic programs of exhaustion constitute a means for T cells to survive in the context of continuous antigen exposure while maintaining limited effector functions such as granzyme B expression and modest interferon (IFN)-γ production (2). Exhausted CD8 T cells (TEX) present a distinct transcriptional program from memory or effector T cells, and a unique epigenetic landscape that becomes largely irreversible over time (3–5). TEX are characterized by abundant expression of the transcription factor TOX, which drives the epigenetic program required for TEX development and maintenance (6–8), and sustain high expression of multiple inhibitory receptors that restrain T cell function (1). Targeting inhibitory receptors on CD8 TEX represents a therapeutic opportunity to reinvigorate anti-tumor immunity. Most prominently, therapies that block the programmed cell death-1 (PD-1) inhibitory pathway have shown unprecedented clinical responses in different tumor types (9) and combination therapies that block additional inhibitory receptors, such as lymphocyte-activation gene-3 (LAG-3) and cytotoxic T-lymphocyte antigen-4 (CTLA-4), can further improve response rates (10, 11).
Yet, it is important to highlight that inhibitory receptors enable TEX survival and are not a unique feature of TEX. Upon activation, effector CD8 T cells rapidly upregulate an array of inhibitory molecules, including PD-1, LAG-3, and CTLA-4. Many of these receptors are directly induced by T cell receptor signaling, constituting a negative feedback loop mechanism that limits T cell-inflicted immunopathology (2). Supporting this conceptual framework, murine autoreactive CD8 T cells were recently found to be restrained by an exhaustion-like program under control of the inhibitory receptor LAG-3 (12), whereas the presence of an “exhausted” gene signature was predictive of a better clinical prognosis in multiple autoimmune disorders (13). These studies suggest that therapeutic induction of an exhaustion-like state may generally be beneficial to prevent tissue damage, but also underline the delicate balance between effective and detrimental responses, best exemplified by autoreactivity triggered by TEX-reinvigorating cancer therapies (1).
Even though CD8 T cell dysfunction has historically been observed in tumor settings, many aspects of our current mechanistic understanding of T cell exhaustion stem from experimental work in mice chronically infected with lymphocytic choriomeningitis virus (LCMV). Elegant studies have confirmed that the phenotypical and transcriptional profiles of tumor-infiltrating lymphocytes (TILs) broadly overlap with exhausted CD8 T cells in chronic LCMV infection (14). Nevertheless, there can be major differences in TEX that have adapted to different microenvironments: chronic inflammation, hypoxic environments, and nutrient restrictions all impose substantial challenges that enhance T cell dysfunction (15). Metabolic alterations, in particular resulting in mitochondrial dysfunction, accelerate and exacerbate the development of exhaustion (16, 17). Therefore, characterization of cancer-associated CD8 TEX dysfunction across species and in different tumors has been complicated by a lack of common defined degrees of exhaustion, as well as functional and phenotypical correlates that correspond to each degree. Longitudinal analysis, as well as functional and metabolic assays and epigenetic mapping, will therefore be key to improve assessment of T cell exhaustion in cancer.
At the population level, T cell exhaustion has been described as a progressive process in which T cells become more dysfunctional over time. Even though mouse models enable a comprehensive assessment of TILs, these analyses are temporally restricted – given that most murine tumors reach endpoint in about one month. In contrast, patients are often only diagnosed when tumors have attained a sizable stage, and tumorigenesis may have been initiated months to years before, thus increasing the likelihood of more pronounced exhaustion. Therefore, differences in time scale for T cell differentiation in mouse tumor models and patients need to be carefully considered. Additional factors that may affect T cell function, such as history of infections, diabetes, and obesity, require stringent characterization in animal models to provide mechanistic insights into potentially confounding effects on anti-tumor immunity. Finally, TIL diversity in patients is caused not only by the presence of non-tumor-specific bystander T cells, but also due to tumor heterogeneity (with regards to driver mutations, availability of nutrients, immune cell infiltrates) and asynchronous generation of neoantigens (with different affinities and expression), culminating in a mixed pool of exhausted PD-1hi CD8 T cells at various dysfunctional stages with different functionalities.
Characterizing heterogeneity among CD8 TEX
Despite heterogeneity among CD8 TEX, all TEX express PD-1 and TOX and share additional adaptations in response to chronic antigen stimulation. Perhaps the most striking finding regarding CD8 TEX diversity was the identification of progenitor exhausted cells (TPEX) which arise early after T cell activation and give rise to other TEX subsets (18–20) (Fig. 1). TPEX are characterized by preserved expression of T cell factor 1 (TCF-1, encoded by the Tcf7 gene), a key transcription factor also found in naïve and memory T cells. Previous studies have shown that TPEX are essential for CD8 TEX maintenance through self-renewal, as well as through differentiation into TCF-1negCD39+ TEX that exert effector/cytotoxic activity (18–20). TPEX express numerous costimulatory molecules, such as CD28, Inducible T-cell costimulator (ICOS), and CD226, while lacking co-expression of many receptors characteristic of more differentiated TEX subsets, such as T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), CD39, LAG-3, and CTLA-4.
Figure 1. Dynamics of exhausted CD8 T cells.

During chronic antigen exposure, PD-1+ TCF-1+ CD8 T cells (TPEX) continuously supply the pool of exhausted T cells (TEX) by self-renewal and conversion into more differentiated PD-1+CD39+TCF-1neg effector-like cells, which express IFN-γ and Granzyme B (GzmB) as well as terminal TEX, which retain GzmB expression. Upon blockade of PD-1/PD-L1 interactions, effective reinvigoration of TEX requires re-priming of TPEX by APCs that provide T cell receptor (TCR) stimulation as well as costimulatory signals through CD28 resulting in an increase in number and function of effector-like TEX. CD4 T cells may directly and indirectly enhance this process through secretion of cytokines such as IL-2 and IL-21 as well as through CD40/CD40L-mediated APC activation. CREDIT: ASHLEY MASTIN/SCIENCE TRANSLATIONAL MEDICINE
Notably, although PD-1/programmed cell death ligand-1 (PD-L1) blockade may directly enhance effector function in differentiated TEX, several murine studies have shown that TPEX are responsible for immunotherapy efficacy by giving rise to the expanded effector pool (14, 18). Upon PD-1/PD-L1 blockade, TPEX proliferate and differentiate into effector-like TEX with enhanced cytotoxic capacity and ability to secrete cytokines, such as IFN-γ. Although less potent than bona fide effector T cells generated in the wake of acute infections or vaccination, effector-like TEX have been shown to be central in controlling viral load in murine LCMV chronic infection (21, 22) as well as tumor growth in B16F10 melanomas (21). Effector-like TEX eventually transition to terminally differentiated TEX, with higher expression of inhibitory receptors and limited ability to produce cytokines and proliferate (although granzyme B expression is preserved). The signals that drive TPEX differentiation and whether TPEX can directly give rise to terminally differentiated TEX depending on cellular interactions and molecular cues still need to be determined (Fig. 1). Importantly, dynamic changes in the frequency of effector-like and terminally differentiated TEX subsets underlie, at least in part, the observation that the functionality of TEX decreases over time.
Additional TEX states or subsets have been proposed by recent studies in chronic LCMV infection. Tsui et al. further pinpointed CD62L-expressing TPEX with a unique transcriptional program and dependent on the transcription factor MYB to be particularly endowed with long-term proliferative and self-renewing properties (23). Another report highlighted that TPEX transition from an early active and less differentiated state to a more quiescent status that is irreversibly committed to exhaustion; this study also identified a new subset of TEX characterized by expression of receptors usually associated with natural killer cells (5). These new studies show that TEX diversity is not fully resolved and continues to be dissected. Future studies in both animal models and patients will need to further investigate these TEX subsets and states and our proposed model for the dynamics of TEX will have to be updated accordingly.
High-throughput profiling techniques. such as single-cell RNA sequencing. have revolutionized the characterization of the tumor microenvironment by highlighting human TIL diversity beyond a handful of markers. Remarkably, despite differences between studies, overall similar populations of human T cells showing signs of chronic antigen stimulation and clonal expansion have been identified across various cancer types (24–26). In patient tumors, in addition to an “exhaustion signature”, CXCL13 expression identifies tumor-specific CD4 and CD8 T cells (27, 28). Furthermore, most studies have described a “pre-dysfunctional” or “transitional” subset of human CD8 TEX corresponding to effector-like TEX, marked by intermediate expression of inhibitory receptors as well as GZMK expression. In addition, TEX cells displaying abundant expression of multiple inhibitory receptors (LAG3, ENTPD1, CTLA4), matching the terminally-differentiated, more dysfunctional cells observed in mice, have also been identified in tumor tissues of patients (24–26, 29). Although many earlier reports did not report human TILs equivalent to the TPEX subset, more recent studies have been able to identify this population in patients with cancer, either by analysis of large data sets (24, 29) or by focusing on antigen-specific CD8 T cells (30). In silico trajectory, T cell receptor sequencing analyses, and even proliferation assays have revealed relationships between different states of human TEX consistent with studies in mouse models (24, 29, 30). Yet, an integrated characterization of TEX phenotype and function in conjunction with clinical data, as well as additional analysis of both the tumor microenvironment and immune cell composition, is still needed to better understand the dynamics and function of TEX subsets in patients with cancer.
Re-priming for reinvigoration of anti-tumor T cell responses
When T cell exhaustion was first described, the primary focus was on reversing this process. Now, novel strategies to improve T cell function during chronic stimulation need to reconcile that epigenetic modifications and metabolic dysregulation can restrict TEX differentiation and functional potential, but also that, among TEX, the small TPEX subset can give rise to cells that retain some functionality. In the last few years, it has become evident that merely blocking individual inhibitory signals might not be sufficient to induce long-lasting, effective anti-tumor responses in a majority of patients with cancer. For instance, we previously demonstrated that CD28-mediated co-stimulation is essential for the proliferation of PD-1+ CD8 T cells and successful anti-PD-1/PD-L1 therapy (31). Recent studies in patients have further pointed out the role of professional antigen-presenting cells (APCs) as well as cellular interactions to promote anti-tumor CD8 T cell responses (29, 32, 33). Together, these data provide a paradigm shift in our understanding of how to reinvigorate dysfunctional T cell responses, implying that this process requires re-priming of tumor-specific TPEX. Even though many aspects of these re-priming events share similarities with the activation of naïve T cells, major differences need to be considered with regards to expression of chemokines and cytokines, costimulatory and inhibitory receptors, and the epigenetic landscape of TPEX. Therefore, combination therapies based on TPEX biology and requirements for differentiation into effector-like TEX would represent novel promising approaches.
TPEX express high XCL-1 (lymphotactin), a chemoattractant for XCR1+ dendritic cells (DC1s) (18). In murine models, consistent with DC1 colocalization, TPEX are mostly found in the T cell zones of secondary lymphoid organs. Likewise, in patients with cancer, TPEX were identified within intratumoral niches containing major histocompatibility complex (MHC)-IIhi APCs, and the presence of these niches in kidney tumors correlated with improved progression-free survival (32). Moreover, it has been shown that APCs in ovarian tumors can provide adequate in situ CD28 costimulation to T cells during PD-1 blockade therapy (33). Finally, our recent observations in patients with hepatocellular carcinoma treated with anti-PD-1 show that, in responders, TPEX are in close proximity to DCs enriched in maturation and regulatory molecules (“mregDC”) (34) and CXCL13+ CD4 T cells, suggesting that this triad in the tumor microenvironment may enhance the clinical response to PD-1 blockade (29).
In murine models, CD4 T cells play a key role for effector-like TEX differentiation (21). Studies of TILs from patients have recently highlighted the presence of tumor-specific CD4 T cells with features of chronic antigen exposure and similarities to follicular helper CD4 T cells (24, 29). CD4 T cells may enhance the function of TEX by providing cytokines such as IL-2 and IL-21 or by activation of APCs (Fig. 1) in a similar manner as described for CD4 help during priming of naïve CD8 T cells. Therefore, it is possible that specific cell interactions provide an adequate environment to support TPEX survival, ensuring TPEX self-renewal and, upon PD-1/PD-L1 blockade, differentiation into effector-like TEX. Hence, intratumoral re-priming of TPEX might play a key role in clinical responses to immunotherapy. In sum, the general focus has recently shifted away from reverting T cell exhaustion to instead focus on better understanding of how interventions modulate the dynamics and function of specific TEX subsets. In addition to optimized strategies to promote TPEX differentiation into effector-like TEX, clinical efficacy may also rely on enhanced survival and function of effector-like TEX. Furthermore, recruitment of TPEX and naïve clones from draining lymph nodes may also need to be harnessed to potentiate responses, especially when tumors lack APC-rich niches.
Additional studies are required to better address these pending questions, which are central to guiding successful combination therapies. Multiple clinical trials are currently underway evaluating the role of cytokines and other agents that may promote APC recruitment and activation or that can bypass the need for some of these cellular interactions by directly activating TEX. The impact of local cues for reinvigoration will also need to be carefully studied. In sum, we must acknowledge that tumor-specific T cells have adapted to chronic antigen stimulation and are therefore exhausted. Furthermore, in cancer, TEX may show additional dysfunctional features due to adaptations to the specific tumor microenvironment, such as nutrient limitations and interactions with suppressive immune cells. Despite differences across diseases and patients, we need to find common patterns to better understand the biology of T cells that respond to persistent antigen. We now possess unmatched tools to comprehensively characterize TEX; appreciating both the limitations and strengths of animal models as well as an extended analysis of patient samples will allow for more efficient progress in the development of novel therapeutic strategies against cancer.
Acknowledgments
Funding:
A.O.K., V.v.d.H., and E.H. were supported by R01 AI153363.
References
- 1.Collier JL, Weiss SA, Pauken KE, Sen DR, Sharpe AH, Not-so-opposite ends of the spectrum: CD8(+) T cell dysfunction across chronic infection, cancer and autoimmunity. Nat Immunol 22, 809–819 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Speiser DE et al. , T cell differentiation in chronic infection and cancer: functional adaptation or exhaustion? Nat Rev Immunol 14, 768–774 (2014). [DOI] [PubMed] [Google Scholar]
- 3.Schietinger A et al. , Tumor-Specific T Cell Dysfunction Is a Dynamic Antigen-Driven Differentiation Program Initiated Early during Tumorigenesis. Immunity 45, 389–401 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Abdel-Hakeem MS et al. , Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat Immunol 22, 1008–1019 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Giles JR et al. , Longitudinal single cell transcriptional and epigenetic mapping of effector, memory, and exhausted CD8 T cells reveals shared biological circuits across distinct cell fates. bioRxiv, (2022). [Google Scholar]
- 6.Khan O et al. , TOX transcriptionally and epigenetically programs CD8. Nature 571, 211–218 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Scott AC et al. , TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Alfei F et al. , TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019). [DOI] [PubMed] [Google Scholar]
- 9.Ribas A, Wolchok JD, Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sharma P et al. , The Next Decade of Immune Checkpoint Therapy. Cancer Discov 11, 838–857 (2021). [DOI] [PubMed] [Google Scholar]
- 11.Andrews LP et al. , Molecular Pathways and Mechanisms of LAG-3 in Cancer Therapy. Clin Cancer Res, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Grebinoski S et al. , Autoreactive CD8(+) T cells are restrained by an exhaustion-like program that is maintained by LAG3. Nat Immunol 23, 868–877 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.McKinney EF, Lee JC, Jayne DR, Lyons PA, Smith KG, T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Miller BC et al. , Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 20, 326–336 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhao S, Peralta RM, Avina-Ochoa N, Delgoffe GM, Kaech SM, Metabolic regulation of T cells in the tumor microenvironment by nutrient availability and diet. Semin Immunol 52, 101485 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.McKinney EF, Smith KGC, Metabolic exhaustion in infection, cancer and autoimmunity. Nat Immunol 19, 213–221 (2018). [DOI] [PubMed] [Google Scholar]
- 17.Yu YR et al. , Disturbed mitochondrial dynamics in CD8(+) TILs reinforce T cell exhaustion. Nat Immunol 21, 1540–1551 (2020). [DOI] [PubMed] [Google Scholar]
- 18.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]
- 19.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]
- 20.Wu T et al. , The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci Immunol 1, (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zander R et al. , CD4(+) T Cell Help Is Required for the Formation of a Cytolytic CD8(+) T Cell Subset that Protects against Chronic Infection and Cancer. Immunity 51, 1028–1042 e1024 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hudson WH et al. , Proliferating Transitory T Cells with an Effector-like Transcriptional Signature Emerge from PD-1. Immunity 51, 1043–1058.e1044 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tsui C et al. , MYB orchestrates T cell exhaustion and response to checkpoint inhibition. Nature 609, 354–360 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zheng L et al. , Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021). [DOI] [PubMed] [Google Scholar]
- 25.Li H et al. , Dysfunctional CD8 T Cells Form a Proliferative, Dynamically Regulated Compartment within Human Melanoma. Cell 176, 775–789 e718 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.van der Leun AM, Thommen DS, Schumacher TN, CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer 20, 218–232 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Liu B, Zhang Y, Wang D, Hu X, Zhang Z, Single-cell meta-analyses reveal responses of tumor-reactive CXCL13(+) T cells to immune-checkpoint blockade. Nat Cancer 3, 1123–1136 (2022). [DOI] [PubMed] [Google Scholar]
- 28.Lowery FJ et al. , Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Magen A et al. , Intratumoral mregDC and CXCL13 T helper niches enable local differentiation of CD8 T cells following PD-1 blockade. bioRxiv, (2022). [Google Scholar]
- 30.Eberhardt CS et al. , Functional HPV-specific PD-1(+) stem-like CD8 T cells in head and neck cancer. Nature 597, 279–284 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kamphorst AO et al. , Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355, 1423–1427 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Jansen CS et al. , An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Duraiswamy J et al. , Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. Cancer Cell 39, 1623–1642 e1620 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Maier B et al. , A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 580, 257–262 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
