CD8+T Cell Exhaustion and Immune Checkpoint Blockade Treatment
DOI:
https://doi.org/10.62051/ppc1bt59Keywords:
Tumor microenvironment (TME); CD8+ T cell exhaustion; cancer immunotherapy; intervention method; immune checkpoint blockade (ICB).Abstract
T cells play an important role in anti-tumor immunity. Its subset that contains the surface receptor CD8, also called cytotoxic T cells, functions to facilitate tumor cell elimination. Chronic exposure of CD8+ T cells to antigens and inflammation can cause the T cells to lose their functions as well as changes in their surface expressions, named T cell exhaustion (Tex). This article reviews the molecular mechanisms of this cellular process and the influence of the tumor microenvironment (TME) on it. This article focuses on the mechanism of action of the four major upregulated immune checkpoint proteins (ICPs) and introduces ICB therapies targeting each ICP. Based on these mechanisms, scientists have suggested various interventions to reverse T-cell exhaustion. Recent clinical data show that although ICB therapy has significant efficacy in some patients, its limitations in patient selection limit the popularization of the treatment effect. ICB which curbs Tex by blocking the signal pathways that suppress T cell functions, summarizes the current understandings and progress, and identifies the gaps on this topic.
Downloads
References
[1] Wang, Q., Qin, Y. & Li, B. CD8+ T cell exhaustion and cancer immunotherapy. Cancer Lett, 2023, 559.
[2] Reina-Campos, M., Scharping, N. E. & Goldrath, A. W. CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol, 2021, 21: 718–738.
[3] Pauken, K. E. & Wherry, E. J. Overcoming T cell exhaustion in infection and cancer. Trends Immunol, 2015, 36: 265–276.
[4] Huang, Y., Jia, A., Wang, Y. & Liu, G. CD8+ T cell exhaustion in anti-tumour immunity: The new insights for cancer immunotherapy. Immunology, 2023, 168: 30–48.
[5] Philip, M. & Schietinger, A. CD8+ T cell differentiation and dysfunction in cancer. Nat Rev Immunol, 2022, 22: 209–223.
[6] Franco, F., Jaccard, A., Romero, P., Yu, Y. R. & Ho, P. C. Metabolic and epigenetic regulation of T-cell exhaustion. Nat Metab, 2020, 2: 1001–1012.
[7] Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med, 2018, 24: 541–550.
[8] Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature, 2019, 571: 211–218.
[9] Wang, Y. et al. Cholesterol-Lowering Intervention Decreases mTOR Complex 2 Signaling and Enhances Antitumor Immunity. Clinical Cancer Research, 2022, 28: 414–424.
[10] Byun, D. J., Wolchok, J. D., Rosenberg, L. M. & Girotra, M. Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol, 2017, 13: 195–207.
[11] Sharma, P. et al. Immune checkpoint therapy—current perspectives and future directions. Cell, 2023, 186: 1652–1669.
[12] Kalia, V., Penny, L. A., Yuzefpolskiy, Y., Baumann, F. M. & Sarkar, S. Quiescence of Memory CD8+ T Cells Is Mediated by Regulatory T Cells through Inhibitory Receptor CTLA-4. Immunity, 2015, 42: 1116–1129.
[13] Martínez-Lostao, L., Anel, A. & Pardo, J. How Do Cytotoxic Lymphocytes Kill Cancer Cells?. Clinical Cancer Research, 2015, 21: 5047–5056.
[14] Saleh, R., Toor, S. M. & Elkord, E. Targeting TIM-3 in solid tumors: innovations in the preclinical and translational realm and therapeutic potential. Expert Opin Ther Targets, 2020, 24: 1251–1262.
[15] Acharya, N., Acharya, N., Sabatos-Peyton, C., Anderson, A. C. & Anderson, A. C. Tim-3 finds its place in the cancer immunotherapy landscape. J Immunother Cancer, 2020, 8.
[16] Ruffo, E., Wu, R. C., Bruno, T. C., Workman, C. J. & Vignali, D. A. A. Lymphocyte-activation gene 3 (LAG3): The next immune checkpoint receptor. Semin Immunol, 2019, 42.
[17] Maruhashi, T., Sugiura, D., Okazaki, I. M. & Okazaki, T. LAG-3: from molecular functions to clinical applications. J Immunother Cancer, 2020, 8.
[18] Dolina, J. S., Van Braeckel-Budimir, N., Thomas, G. D. & Salek-Ardakani, S. CD8+ T Cell Exhaustion in Cancer. Front Immunol, 2021, 12.
[19] Gaikwad, S., Agrawal, M. Y., Kaushik, I., Ramachandran, S. & Srivastava, S. K. Immune checkpoint proteins: Signaling mechanisms and molecular interactions in cancer immunotherapy. Semin Cancer Biol, 2022, 86: 137–150.
[20] Zhang, H. et al. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. Journal of Experimental and Clinical Cancer Research, 2021, 40.
[21] Kozłowski, M., Borzyszkowska, D. & Cymbaluk-Płoska, A. The Role of TIM-3 and LAG-3 in the Microenvironment and Immunotherapy of Ovarian Cancer. Biomedicines, 2022, 10.
[22] Sauer, N. et al. TIM-3 as a promising target for cancer immunotherapy in a wide range of tumors. Cancer Immunology, Immunotherapy, 2023, 72: 3405–3425.
[23] Aggarwal, V., Workman, C. J. & Vignali, D. A. A. LAG-3 as the third checkpoint inhibitor. Nat Immunol, 2023, 24: 1415–1422.
[24] Joller, N., Anderson, A. C. & Kuchroo, V. K. LAG-3, TIM-3, and TIGIT: Distinct functions in immune regulation. Immunity, 2024, 57: 206–222.
[25] Yi, M. et al. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Molecular Cancer, 2022, 21.
[26] Zhang, H. et al. Roles of tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for solid cancers. Molecular Cancer, 2023, 22.
[27] Liu, B. et al. Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat Cancer, 2022, 3: 108–121.
Downloads
Published
Conference Proceedings Volume
Section
License

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.