Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Viewpoint
  • Published:

Defining ‘T cell exhaustion’

Abstract

‘T cell exhaustion’ is a broad term that has been used to describe the response of T cells to chronic antigen stimulation, first in the setting of chronic viral infection but more recently in response to tumours. Understanding the features of and pathways to exhaustion has crucial implications for the success of checkpoint blockade and adoptive T cell transfer therapies. In this Viewpoint article, 18 experts in the field tell us what exhaustion means to them, ranging from complete lack of effector function to altered functionality to prevent immunopathology, with potential differences between cancer and chronic infection. Their responses highlight the dichotomy between terminally differentiated exhausted T cells that are TCF1 and the self-renewing TCF1+ population from which they derive. These TCF1+ cells are considered by some to have stem cell-like properties akin to memory T cell populations, but the developmental relationships are unclear at present. Recent studies have also highlighted an important role for the transcriptional regulator TOX in driving the epigenetic enforcement of exhaustion, but key questions remain about the potential to reverse the epigenetic programme of exhaustion and how this might affect the persistence of T cell populations.

The contributors

Christian U. Blank is a medical oncologist and principal investigator at the Netherlands Cancer Institute. He is Professor of Haematology/Oncology at the University of Regensburg, Germany, and received an MBA degree from the University of Warwick, UK. His research interests include neoadjuvant immunotherapies, targeted and biological response modifiers, and prognostic markers for cancer immunotherapies.

W. Nicholas Haining is a physician–scientist and Vice-President for Discovery Oncology and Immunology at Merck Research Laboratories. His former academic laboratory at the Dana-Farber Cancer Institute and the Broad Institute focused on understanding the transcriptional control of T cell exhaustion and on identifying regulators of the immune response to cancer in tumour and immune cells.

Werner Held’s laboratory has a long-standing interest in understanding the development, differentiation and function of natural killer cells and CD8+ T cells. Current work focuses on CD8+ T cell differentiation in response to acute and chronic infections as well as cancer.

Patrick G. Hogan’s research centres on mechanisms and regulation of cellular calcium signalling, the biology of the nuclear factor of activated T cells (NFAT) family of transcription factors and the transcriptional control of immune cell development and function.

Axel Kallies is a professor at the University of Melbourne, Australia. His laboratory studies the molecular control of CD8+ cytotoxic T cell and regulatory T cell differentiation with a focus on populations residing in non-lymphoid tissue, including healthy tissues and tumours. The Kallies laboratory has developed and applied genetic and molecular approaches to this field, including novel gene reporters, metabolic techniques, transcriptional profiling, chromatin immunoprecipitation and accessible chromatin sequencing.

Enrico Lugli’s laboratory is focused on understanding the biological mechanisms at the basis of memory T cell responses and homeostasis in humans and how this information can be exploited to favour antitumour immune responses in patients with cancer. The group is specialized in single-cell technologies, in particular high-dimensional flow cytometry.

Rachel C. Lynn is an associate director of research at Lyell Immunopharma. She received her PhD degree from the the University of Pennsylvania, where she developed multiple preclinical chimeric antigen receptor (CAR) T cell therapy platforms. During her postdoctoral work with Crystal Mackall at Stanford University, she developed models to interrogate and strategies to mitigate CAR T cell exhaustion. At Lyell Immunopharma, her research group will continue to investigate optimal strategies for adoptive T cell therapy in cancer.

Mary Philip is an assistant professor in the Department of Medicine, Division of Hematology and Oncology and Department of Pathology, Microbiology, and Immunology at Vanderbilt University Medical Center. She did her haematology and oncology fellowship training at the Fred Hutchinson Cancer Research Center and the University of Washington and then joined Andrea Schietinger’s group at Memorial Sloan Kettering Cancer Center investigating the epigenetic and transcriptional regulation of T cell dysfunction and reprogrammability in cancer. She is a physician–scientist with clinical expertise in haematological cancers, and her research group at Vanderbilt University Medical Center is developing innovative models to dissect the regulation of T cell dysfunction in cancer and T cell lymphomas.

Anjana Rao’s laboratory studies transcriptional regulation in several cell types, including CD4+ and CD8+ T cells. One current focus is elucidation of the transcriptional and epigenetic regulatory mechanisms operating in T cells and other immune cells within the tumour microenvironment.

Nicholas P. Restifo was trained at Memorial Sloan Kettering Cancer Center and was a principal investigator at the National Cancer Institute. His work has focused on the use of adoptively transferred T cells in immunotherapy for cancer for the past 31 years. He recently joined Lyell Immunopharma.

Andrea Schietinger is an assistant member in the Immunology Program at Memorial Sloan Kettering Cancer Center. During her PhD studies with Hans Schreiber at the University of Chicago, she studied how aberrant glycosylation of wild-type proteins in cancer cells creates tumour-specific neoantigens. As a postdoctoral fellow in Philip Greenberg’s laboratory at the University of Washington in Seattle, she defined the transcriptional programmes associated with T cell self-tolerance and tumour-specific T cell dysfunction. Her laboratory at Memorial Sloan Kettering Cancer Center studies the regulatory molecular and epigenetic mechanisms underlying T cell differentiation and dysfunction in the context of self-tolerance, autoimmunity and tumours.

Ton N. Schumacher is a principal investigator at the Netherlands Cancer Institute and Professor of Immunotechnology at Leiden University. His research focuses on the dissection of tumour-specific T cell responses in human cancer. In addition to his academic role, he is the founder of four biotech companies and a venture partner at Third Rock Ventures.

Pamela L. Schwartzberg is a senior investigator at the National Institute of Allergy and Infectious Diseases. She completed her MD and PhD degrees at Columbia University and her postdoctoral studies at the National Cancer Institute. Her laboratory studies signalling pathways in T cells using genetic, cellular, biochemical and genomic approaches to understand how these affect responses to infection and immunization with a focus on molecules and pathways affected by primary immunodeficiencies.

Arlene H. Sharpe is Chair of the Department of Immunology at Harvard Medical School. Her laboratory focuses on understanding the diverse roles of costimulatory and coinhibitory signals in infection, autoimmunity and cancer.

Daniel E. Speiser’s team has investigated and treated cancer patients with immunotherapy for the past 30 years. The demonstration and mechanistic elucidation of graft-versus-leukaemia effects and autologous T cell reactivity have helped to pave the way for remarkable therapy innovations in oncology.

E. John Wherry is the Barbara and Richard Schiffrin President’s Distinguished Professor, Chair of the Department of Systems Pharmacology and Translational Therapeutics in the Perelman School of Medicine and Director of the Penn Institute for Immunology. His research has focused on T cell exhaustion in chronic viral infections and cancer. His work has defined the developmental biology and molecular regulation of T cell exhaustion and used this information to inform immunotherapy for human cancer.

Benjamin A. Youngblood is an associate member in the Department of Immunology at St Jude Children’s Research Hospital. His postdoctoral training in Rafi Ahmed’s laboratory focused on the epigenetic regulation of memory CD8+ T cell differentiation. In 2014, he joined the faculty at St Jude Children’s Research Hospital and has developed a research programme studying epigenetic mechanisms that regulate the development of functional and non-functional CD8+ T cells during viral infection, cancer and autoimmunity.

Dietmar Zehn is a full professor and Chair of the Division of Animal Physiology and Immunology of the Technical University of Munich. His research focuses on understanding the molecular and cellular mechanisms that control the differentiation of T cells in acute and chronic infection, with specific interests in T cell exhaustion and T cell receptor signal strength.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The knowns and unknowns of T cell exhaustion.

Similar content being viewed by others

References

  1. Speiser, D. E. et al. T cell differentiation in chronic infection and cancer: functional adaptation or exhaustion? Nat. Rev. Immunol. 14, 768–774 (2014).

    CAS  PubMed  Google Scholar 

  2. Jin, X. et al. Dramatic rise in plasma viremia after CD8+ T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 189, 991–998 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Wherry, E. J. T cell exhaustion. Nat. Immunol. 12, 492–499 (2011).

    CAS  PubMed  Google Scholar 

  4. McLane, L. M., Abdel-Hakeem, M. S. & Wherry, E. J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 37, 457–495 (2019).

    CAS  PubMed  Google Scholar 

  5. Martinez, G. J. et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Yao, C. et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 20, 890–901 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    CAS  PubMed  PubMed Central  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).

    CAS  PubMed  Google Scholar 

  9. Seo, H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410–12415 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, J. et al. Nr4a transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Man, K. et al. Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity 47, 1129–1141 (2017).

    CAS  PubMed  Google Scholar 

  13. Macian, F. et al. Transcriptional mechanisms underlying lymphocyte tolerance. Cell 109, 719–731 (2002).

    CAS  PubMed  Google Scholar 

  14. Heissmeyer, V. et al. Calcineurin imposes T cell unresponsiveness through targeted proteolysis of signaling proteins. Nat. Immunol. 5, 255–265 (2004).

    CAS  PubMed  Google Scholar 

  15. Philip, M. & Schietinger, A. Heterogeneity and fate choice: T cell exhaustion in cancer and chronic infections. Curr. Opin. Immunol. 58, 98–103 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789 (2019).

    CAS  PubMed  Google Scholar 

  17. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Thommen, D. S. et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat. Med. 24, 994–1004 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Scheper, W. et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat. Med. 25, 89–94 (2019).

    CAS  PubMed  Google Scholar 

  20. Simoni, Y. et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018).

    CAS  PubMed  Google Scholar 

  21. Wherry, E. J. et al. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77, 4911–4927 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zajac, A. J. et al. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205–2213 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Youngblood, B. et al. Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells. Immunity 35, 400–412 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Ahrends, T. et al. CD4+ T cell help confers a cytotoxic T cell effector program including coinhibitory receptor downregulation and increased tissue invasiveness. Immunity 47, 848–861 (2017).

    CAS  PubMed  Google Scholar 

  26. Akbar, A. N. & Henson, S. M. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat. Rev. Immunol. 11, 289–295 (2011).

    CAS  PubMed  Google Scholar 

  27. Angelosanto, J. M., Blackburn, S. D., Crawford, A. & Wherry, E. J. Progressive loss of memory T cell potential and commitment to exhaustion during chronic viral infection. J. Virol. 86, 8161–8170 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Paley, M. A. et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Utzschneider, D. T. 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).

    CAS  PubMed  Google Scholar 

  31. Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211.e10 (2019).

    CAS  PubMed  Google Scholar 

  32. Brummelman, J. et al. High-dimensional single cell analysis identifies stem-like cytotoxic CD8+ T cells infiltrating human tumors. J. Exp. Med. 215, 2520–2535 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Jeannet, G. et al. Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8 T cell memory. Proc. Natl Acad. Sci. USA 107, 9777–9782 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Wu, T. et al. The TCF1–Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci. Immunol. 1, eaai8593 (2016).

    PubMed  PubMed Central  Google Scholar 

  35. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1CD8+ tumor-infiltrating T cells. Immunity 50, 181–194 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Jadhav, R. R. et al. Epigenetic signature of PD-1+TCF1+ CD8 T cells that act as resource cells during chronic viral infection and respond to PD-1 blockade. Proc. Natl Acad. Sci. USA 116, 14113–14118 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Bengsch, B. et al. Epigenomic-guided mass cytometry profiling reveals disease-specific features of exhausted CD8 T cells. Immunity 48, 1029–1045 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Gattinoni, L. et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 15, 808–813 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Gattinoni, L. et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Leong, Y. A. et al. CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat. Immunol. 17, 1187–1196 (2016).

    CAS  PubMed  Google Scholar 

  43. Pauken, K. E. et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Philip, M. et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545, 452–456 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Liu, X. et al. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature 567, 525–529 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Wang, X. et al. TOX promotes the exhaustion of antitumor CD8+ T cells by preventing PD1 degradation in hepatocellular carcinoma. J. Hepatol. https://doi.org/10.1016/j.jhep.2019.05.015 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Scott-Browne, J. P. et al. Dynamic changes in chromatin accessibility occur in CD8+ T cells responding to viral infection. Immunity 45, 1327–1340 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Mognol, G. et al. Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells. Proc. Natl Acad. Sci. USA 114, E2776–E2785 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Blackburn, S. D., Shin, H., Freeman, G. J. & Wherry, E. J. Selective expansion of a subset of exhausted CD8 T cells by αPD-L1 blockade. Proc. Natl Acad. Sci. USA 105, 15016–15021 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Ghoneim, H. E. et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142–157 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Odorizzi, P. M. et al. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J. Exp. Med. 212, 1125–1137 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Teijaro, J. R. et al. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340, 207–211 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Stephen, T. L. et al. SATB1 expression governs epigenetic repression of PD-1 in tumor-reactive T cells. Immunity 46, 51–64 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Pilipow, K. et al. Antioxidant metabolism regulates CD8+ T memory stem cell formation and antitumor immunity. JCI Insight 3, 122299 (2018).

    PubMed  Google Scholar 

  56. McKinney, E. F., Lee, J. C., Jayne, D. R., Lyons, P. A. & Smith, K. G. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Utzschneider, D. T. et al. T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat. Immunol. 14, 603–610 (2013).

    CAS  PubMed  Google Scholar 

  58. Willimsky, G. & Blankenstein, T. Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance. Nature 437, 141–146 (2005).

    CAS  PubMed  Google Scholar 

  59. Doedens, A. L. et al. Hypoxia-inducible factors enhance the effector responses of CD8+ T cells to persistent antigen. Nat. Immunol. 14, 1173–1182 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Frebel, H. et al. Programmed death 1 protects from fatal circulatory failure during systemic virus infection of mice. J. Exp. Med. 209, 2485–2499 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Moskophidis, D., Lechner, F., Pircher, H. & Zinkernagel, R. M. Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells. Nature 362, 758–761 (1993).

    CAS  PubMed  Google Scholar 

  62. Rosenberg, S. A. & Restifo, N. P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Vodnala, S. K. et al. T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science 363, eaau0135 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Lynn, R. C. et al. c-Jun overexpressing CAR-T cells are exhaustion-resistant and functionally superior. Nature (in the press).

  65. Overwijk, W. W. et al. Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. J. Exp. Med. 198, 569–580 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

P.G.H. and A.R. are funded by National Institutes of Health grants AI109842, AI040127 and AI084167, and U01 DE028227. A.K. is supported by a fellowship by the National Health and Medical Research Council of Australia. E.L. is supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC IG20607).

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Christian U. Blank, W. Nicholas Haining, Werner Held, Patrick G. Hogan, Axel Kallies, Enrico Lugli, Rachel C. Lynn, Mary Philip, Anjana Rao, Nicholas P. Restifo, Andrea Schietinger, Ton N. Schumacher, Pamela L. Schwartzberg, Arlene H. Sharpe, Daniel E. Speiser, E. John Wherry, Benjamin A. Youngblood or Dietmar Zehn.

Ethics declarations

Competing interests

W.N.H. is an employee of Merck & Co. and a founder of Arsenal Biosciences. The La Jolla Institute for Immunology (LJI) has received a research grant from Lyell lmmunopharma to support aspects of the work of P.G.H. on transcriptional mechanisms in CD8+ T cells. E.L. receives preclinical research funding from Bristol-Myers Squibb. LJI is the recipient of a research grant from Lyell lmmunopharma, which supports studies in the laboratory of A.R. using mouse models to elucidate the transcriptional and epigenetic programmes operating in CD8+ tumour-infiltrating T cells. LJI has a pending patent, PCT/US201B/062354, covering the use and production of engineered immune cells to disrupt NFAT–AP-1 pathway transcription factors, including NR4A1/2/3, TOX and TOX2, with A.R. listed as one of the inventors. LJI is the recipient of a research grant from the Takeda-Sanford Innovation Alliance for research in the laboratory of A.R. related to NR4A in human CD8+ T cells. N.P.R. and R.C.L. are employees of Lyell Immunopharma. A.H.S. has patents on the PD1 pathway licenced by Roche/Genentech and Novartis, consults for Novartis, is on the scientific advisory boards for Surface Oncology, Sqz Biotech, Elstar Therapeutics, Elpiscience, Selecta and Monopteros, and has research funding from Novartis, Roche, Ipsen, UCB, Quark Ventures and Merck. E.J.W. is a member of the Parker Institute for Cancer lmmunotherapy, which supported the present study. E.J.W. has consulting agreements with and/or is on the scientific advisory board for Merck, Roche, Pieris, Elstar and Surface Oncology. E.J.W. has a patent licensing agreement on the PD1 pathway with Roche/Genentech. E.J.W. is a founder of Arsenal Biosciences. B.A.Y. has patents associated with epigenetic programming of T cells. He has received honoraria for speaking at companies (less than US$5,000). The other authors declare no competing interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Blank, C.U., Haining, W.N., Held, W. et al. Defining ‘T cell exhaustion’. Nat Rev Immunol 19, 665–674 (2019). https://doi.org/10.1038/s41577-019-0221-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41577-019-0221-9

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer