Senescent Tumor CD8+ T Cells: Mechanisms of Induction and Challenges to Immunotherapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Exhaustion and Senescence
3. Mechanisms of T Cell Senescence Induction
3.1. Signaling Pathways Involved in Tc Senescence
3.2. The TME Drives Tc Premature Senescence
3.2.1. Immune and Tumor Cells
3.2.2. Metabolic Changes
3.2.3. Chemotherapeutics and Radiation Therapy
3.3. Age-Related Replicative Senescence
4. Tc Cell Senescence and Effects on Immunotherapy Response
4.1. Checkpoint Inhibitors
4.2. CAR-T Cell Therapy
4.3. Targeting T Cell Senescence
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 1–18. [Google Scholar] [CrossRef]
- Martinez-Lostao, L.; Anel, A.; Pardo, J. How Do Cytotoxic Lymphocytes Kill Cancer Cells? Clin. Cancer Res. 2015, 21, 5047–5056. [Google Scholar] [CrossRef] [Green Version]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. New Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
- Barber, D.L.; Wherry, E.J.; Masopust, D.; Zhu, B.; Allison, J.; Sharpe, A.H.; Freeman, G.J.; Ahmed, R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006, 439, 682–687. [Google Scholar] [CrossRef]
- Locke, F.L.; Neelapu, S.S.; Bartlett, N.L.; Siddiqi, T.; Chavez, J.C.; Hosing, C.M.; Ghobadi, A.; Budde, L.E.; Bot, A.; Rossi, J.M.; et al. Phase 1 Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma. Mol. Ther. 2017, 25, 285–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef] [PubMed]
- Vitale, C.; Strati, P. CAR T-Cell Therapy for B-Cell non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia: Clinical Trials and Real-World Experiences. Front. Oncol. 2020, 10, 849. [Google Scholar] [CrossRef] [PubMed]
- Zou, W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat. Rev. Cancer 2005, 5, 263–274. [Google Scholar] [CrossRef]
- Appay, V.; Nixon, D.F.; Donahoe, S.M.; Gillespie, G.M.; Dong, T.; King, A.; Ogg, G.S.; Spiegel, H.M.; Conlon, C.; Spina, C.A.; et al. HIV-Specific Cd8+ T Cells Produce Antiviral Cytokines but Are Impaired in Cytolytic Function. J. Exp. Med. 2000, 192, 63–76. [Google Scholar] [CrossRef] [Green Version]
- Wherry, E.J.; Ha, S.-J.; Kaech, S.M.; Haining, W.N.; Sarkar, S.; Kalia, V.; Subramaniam, S.; Blattman, J.N.; Barber, D.L.; Ahmed, R. Molecular Signature of CD8+ T Cell Exhaustion during Chronic Viral Infection. Immunity 2007, 27, 670–684. [Google Scholar] [CrossRef] [Green Version]
- Zelle-Rieser, C.; Thangavadivel, S.; Biedermann, R.; Brunner, A.; Stoitzner, P.; Willenbacher, E.; Greil, R.; Jöhrer, K. T cells in multiple myeloma display features of exhaustion and senescence at the tumor site. J. Hematol. Oncol. 2016, 9, 116. [Google Scholar] [CrossRef] [Green Version]
- Li, K.-K.; Adams, D.H. Antitumor CD8+ T cells in hepatocellular carcinoma: Present but exhausted. Hepatology 2014, 59, 1232–1234. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Hoft, D.F.; Peng, G. Senescent T cells within suppressive tumor microenvironments: Emerging target for tumor immunotherapy. J. Clin. Investig. 2020, 130, 1073–1083. [Google Scholar] [CrossRef] [PubMed]
- Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Shao, Q.; Peng, G. Exhaustion and senescence: Two crucial dysfunctional states of T cells in the tumor microenvironment. Cell. Mol. Immunol. 2019, 17, 27–35. [Google Scholar] [CrossRef]
- Dolfi, D.V.; Mansfield, K.D.; Polley, A.M.; Doyle, S.A.; Freeman, G.J.; Pircher, H.; Schmader, K.E.; Wherry, E.J. Increased T-bet is associated with senescence of influenza virus-specific CD8 T cells in aged humans. J. Leukoc. Biol. 2013, 93, 825–836. [Google Scholar] [CrossRef] [Green Version]
- Crespo, J.; Sun, H.; Welling, T.H.; Tian, Z.; Zou, W. T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr. Opin. Immunol. 2013, 25, 214–221. [Google Scholar] [CrossRef] [Green Version]
- Bour-Jordan, H.; Esensten, J.H.; Martínez-Llordella, M.; Penaranda, C.; Stumpf, M.; Bluestone, J.A. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/B7 family. Immunol. Rev. 2011, 241, 180–205. [Google Scholar] [CrossRef] [Green Version]
- Papalampros, A.; Vailas, M.; Ntostoglou, K.; Chiloeches, M.L.; Sakellariou, S.; Chouliari, N.V.; Samaras, M.G.; Veltsista, P.D.; Theodorou, S.D.P.; Margetis, A.T.; et al. Unique Spatial Immune Profiling in Pancreatic Ductal Adenocarcinoma with Enrichment of Exhausted and Senescent T Cells and Diffused CD47-SIRPα Expression. Cancers 2020, 12, 1825. [Google Scholar] [CrossRef]
- Li, H.; Wu, K.; Tao, K.; Chen, L.; Zheng, Q.; Lu, X.; Liu, J.; Shi, L.; Liu, C.; Wang, G.; et al. Tim-3/galectin-9 signaling pathway mediates T-cell dysfunction and predicts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma. Hepatology 2012, 56, 1342–1351. [Google Scholar] [CrossRef]
- Gruber, I.; El Yousfi, S.; Dürr-Störzer, S.; Wallwiener, D.; Solomayer, E.F.; Fehm, T. Down-regulation of CD28, TCR-zeta (zeta) and up-regulation of FAS in peripheral cytotoxic T-cells of primary breast cancer patients. Anticancer Res. 2008, 28, 779–784. [Google Scholar] [PubMed]
- Onyema, O.O.; DeCoster, L.; Njemini, R.; Forti, L.N.; Bautmans, I.; De Waele, M.; Mets, T. Chemotherapy-induced changes and immunosenescence of CD8+ T-cells in patients with breast cancer. Anticancer Res. 2015, 35, 1481–1489. [Google Scholar] [PubMed]
- Song, G.; Wang, X.; Jia, J.; Yuan, Y.; Wan, F.; Zhou, X.; Yang, H.; Ren, J.; Gu, J.; Lyerly, H.K. Elevated level of peripheral CD8+CD28− T lymphocytes are an independent predictor of progression-free survival in patients with metastatic breast cancer during the course of chemotherapy. Cancer Immunol. Immunother. 2013, 62, 1123–1130. [Google Scholar] [CrossRef] [PubMed]
- Suen, H.; Brown, R.; Yang, S.; Weatherburn, C.; Ho, P.J.; Woodland, N.; Nassif, N.; Barbaro, P.; Bryant, C.; Hart, D.; et al. Multiple myeloma causes clonal T-cell immunosenescence: Identification of potential novel targets for promoting tumour immunity and implications for checkpoint blockade. Leukemia 2016, 30, 1716–1724. [Google Scholar] [CrossRef] [PubMed]
- Henson, S.M.; Lanna, A.; Riddell, N.; Franzese, O.; Macaulay, R.; Griffiths, S.J.; Puleston, D.J.; Watson, A.S.; Simon, A.K.; Tooze, S.A.; et al. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J. Clin. Investig. 2014, 124, 4004–4016. [Google Scholar] [CrossRef]
- Lanna, A.; Henson, S.M.; Escors, D.; Akbar, A.N. The kinase p38 activated by the metabolic regulator AMPK and scaffold TAB1 drives the senescence of human T cells. Nat. Immunol. 2014, 15, 965–972. [Google Scholar] [CrossRef] [Green Version]
- Lanna, A.; Gomes, D.C.; Müller-Durovic, B.; McDonnell, T.; Escors, D.; Gilroy, D.W.; Lee, J.H.; Karin, M.; Akbar, A.N. A sestrin-dependent Erk–Jnk–p38 MAPK activation complex inhibits immunity during aging. Nat. Immunol. 2017, 18, 354–363. [Google Scholar] [CrossRef] [Green Version]
- Pereira, B.I.; De Maeyer, R.P.H.; Covre, L.P.; Nehar-Belaid, D.; Lanna, A.; Ward, S.; Marches, R.; Chambers, E.S.; Gomes, D.C.O.; Riddell, N.E.; et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nat. Immunol. 2020, 21, 684–694. [Google Scholar] [CrossRef]
- Desdín-Micó, G.; Soto-Heredero, G.; Aranda, J.F.; Oller, J.; Carrasco, E.; Gabandé-Rodríguez, E.; Blanco, E.M.; Alfranca, A.; Cussó, L.; Desco, M.; et al. T cells with dysfunctional mitochondria induce multimorbidity and premature senescence. Science 2020, 368, 1371–1376. [Google Scholar] [CrossRef]
- Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
- Rapp, M.; Wintergerst, M.W.M.; Kunz, W.G.; Vetter, V.K.; Knott, M.M.L.; Lisowski, D.; Haubner, S.; Moder, S.; Thaler, R.; Eiber, S.; et al. CCL22 controls immunity by promoting regulatory T cell communication with dendritic cells in lymph nodes. J. Exp. Med. 2019, 216, 1170–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, J.-H.; Kim, B.S.; Lee, S.-K. Regulatory T Cells in Tumor Microenvironment and Approach for Anticancer Immunotherapy. Immune Netw. 2020, 20, e4. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Jiang, P.; Wei, S.; Xu, X.; Wang, J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 2020, 19, 1–23. [Google Scholar] [CrossRef] [PubMed]
- Ye, J.; Huang, X.; Hsueh, E.C.; Zhang, Q.; Ma, C.; Zhang, Y.; Varvares, M.A.; Hoft, D.F.; Peng, G. Human regulatory T cells induce T-lymphocyte senescence. Blood 2012, 120, 2021–2031. [Google Scholar] [CrossRef]
- Liu, X.; Mo, W.; Ye, J.; Li, L.; Zhang, Y.; Hsueh, E.C.; Hoft, D.F.; Peng, G. Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition. Nat. Commun. 2018, 9, 249. [Google Scholar] [CrossRef]
- Ji, A.L.; Rubin, A.J.; Thrane, K.; Jiang, S.; Reynolds, D.L.; Meyers, R.M.; Guo, M.G.; George, B.M.; Mollbrink, A.; Bergenstråhle, J.; et al. Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell Carcinoma. Cell 2020, 182, 497–514.e22. [Google Scholar] [CrossRef]
- Sakuishi, K.; Ngiow, S.F.; Sullivan, J.M.; Teng, M.W.L.; Kuchroo, V.K.; Smyth, M.J.; Anderson, A.C. TIM3+FOXP3+regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer. OncoImmunology 2013, 2, e23849. [Google Scholar] [CrossRef] [Green Version]
- Zhou, X.; Zhao, S.; He, Y.; Geng, S.; Shi, Y.; Wang, B. Precise Spatiotemporal Interruption of Regulatory T-cell–Mediated CD8+ T-cell Suppression Leads to Tumor Immunity. Cancer Res. 2018, 79, 585–597. [Google Scholar] [CrossRef] [Green Version]
- Hu, C.; Pang, B.; Lin, G.; Zhen, Y.; Yi, H. Energy metabolism manipulates the fate and function of tumour myeloid-derived suppressor cells. Br. J. Cancer 2020, 122, 23–29. [Google Scholar] [CrossRef]
- Montes, C.L.; Chapoval, A.; Nelson, J.A.; Orhue, V.; Zhang, X.; Schulze, D.H.; Strome, S.E.; Gastman, B.R. Tumor-induced senescent T cells with suppressor function: A potential form of tumor immune evasion. Cancer Res. 2008, 68, 870–879. [Google Scholar] [CrossRef] [Green Version]
- Ye, J.; Ma, C.; Hsueh, E.C.; Dou, J.; Mo, W.; Liu, S.; Han, B.; Huang, Y.; Zhang, Y.; Varvares, M.; et al. TLR 8 signaling enhances tumor immunity by preventing tumor-induced T-cell senescence. EMBO Mol. Med. 2014, 6, 1294–1311. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Liu, X.; Sanders, K.L.; Edwards, J.L.; Ye, J.; Si, F.; Gao, A.; Huang, L.; Hsueh, E.C.; Ford, D.A.; et al. TLR8-Mediated Metabolic Control of Human Treg Function: A Mechanistic Target for Cancer Immunotherapy. Cell Metab. 2019, 29, 103–123.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buck, M.D.; O’Sullivan, D.; Pearce, E.L. T cell metabolism drives immunity. J. Exp. Med. 2015, 212, 1345–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Neill, L.A.; Pearce, E.J. Immunometabolism governs dendritic cell and macrophage function. J. Exp. Med. 2015, 213, 15–23. [Google Scholar] [CrossRef] [PubMed]
- Jiang, B. Aerobic glycolysis and high level of lactate in cancer metabolism and microenvironment. Gene Funct. Dis. 2017, 4, 25–27. [Google Scholar] [CrossRef]
- Sukumar, M.; Liu, J.; Ji, Y.; Subramanian, M.; Crompton, J.G.; Yu, Z.; Roychoudhuri, R.; Palmer, D.C.; Muranski, P.; Karoly, E.D.; et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Investig. 2013, 123, 4479–4488. [Google Scholar] [CrossRef]
- Wang, R.; Dillon, C.P.; Shi, L.Z.; Milasta, S.; Carter, R.; Finkelstein, D.; McCormick, L.L.; Fitzgerald, P.; Chi, H.; Munger, J.; et al. The Transcription Factor Myc Controls Metabolic Reprogramming upon T Lymphocyte Activation. Immunity 2011, 35, 871–882. [Google Scholar] [CrossRef] [Green Version]
- Angelin, A.; Gil-de-Gómez, L.; Dahiya, S.; Jiao, J.; Guo, L.; Levine, M.H.; Wang, Z.; Quinn, W.J., III; Kopinski, P.K.; Wang, L.; et al. Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. Cell Metab. 2017, 25, 1282–1293.e7. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, M.K.; Sinha, P.; Clements, V.K.; Rodriguez, P.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010, 70, 68–77. [Google Scholar] [CrossRef] [Green Version]
- Wu, K.; Lin, K.; Li, X.; Yuan, X.; Xu, P.; Ni, P.; Xu, D. Redefining Tumor-Associated Macrophage Subpopulations and Functions in the Tumor Microenvironment. Front. Immunol. 2020, 11, 1731. [Google Scholar] [CrossRef]
- Clayton, A.; Al-Taei, S.; Webber, J.P.; Mason, M.D.; Tabi, Z. Cancer Exosomes Express CD39 and CD73, Which Suppress T Cells through Adenosine Production. J. Immunol. 2011, 187, 676–683. [Google Scholar] [CrossRef]
- Parish, S.T.; Kim, S.; Sekhon, R.K.; Wu, J.E.; Kawakatsu, Y.; Effros, R.B. Adenosine Deaminase Modulation of Telomerase Activity and Replicative Senescence in Human CD8 T Lymphocytes. J. Immunol. 2010, 184, 2847–2854. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.-H.; Choi, S.-Y.; Jung, N.-C.; Song, J.-Y.; Seo, H.G.; Lee, H.S.; Lim, D.-S. The Effect of the Tumor Microenvironment and Tumor-Derived Metabolites on Dendritic Cell Function. J. Cancer 2020, 11, 769–775. [Google Scholar] [CrossRef] [Green Version]
- Vang, T.; Torgersen, K.M.; Sundvold, V.; Saxena, M.; Levy, F.O.; Skålhegg, B.S.; Hansson, V.; Mustelin, T.; Taskén, K. Activation of the COOH-terminal Src Kinase (Csk) by cAMP-dependent Protein Kinase Inhibits Signaling through the T Cell Receptor. J. Exp. Med. 2001, 193, 497–508. [Google Scholar] [CrossRef] [Green Version]
- Bopp, T.; Becker, C.; Klein, M.; Klein-Hessling, S.; Palmetshofer, A.; Serfling, E.; Heib, V.; Becker, M.; Kubach, J.; Schmitt, S.; et al. Cyclic adenosine monophosphate is a key component of regulatory T cell–mediated suppression. J. Exp. Med. 2007, 204, 1303–1310. [Google Scholar] [CrossRef] [Green Version]
- Ustun, C.; Miller, J.S.; Munn, D.H.; Weisdorf, D.J.; Blazar, B.R. Regulatory T cells in acute myelogenous leukemia: Is it time for immunomodulation? Blood 2011, 118, 5084–5095. [Google Scholar] [CrossRef] [Green Version]
- Campisi, J.; d’Adda di Fagagna, D. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef]
- Faheem, M.M.; Seligson, N.D.; Ahmad, S.M.; Rasool, R.U.; Gandhi, S.G.; Bhagat, M.; Goswami, A. Convergence of therapy-induced senescence (TIS) and EMT in multistep carcinogenesis: Current opinions and emerging perspectives. Cell Death Discov. 2020, 6, 1–12. [Google Scholar] [CrossRef]
- Sehl, M.E.; Carroll, J.E.; Horvath, S.; Bower, J.E. The acute effects of adjuvant radiation and chemotherapy on peripheral blood epigenetic age in early stage breast cancer patients. NPJ Breast Cancer 2020, 6, 23. [Google Scholar] [CrossRef]
- Lee, J.-J.; Nam, C.-E.; Cho, S.-H.; Park, K.-S.; Chung, I.-J.; Kim, H.-J. Telomere length shortening in non-Hodgkin’s lymphoma patients undergoing chemotherapy. Ann. Hematol. 2003, 82, 492–495. [Google Scholar] [CrossRef]
- d’Adda di Fagagna, F.; Reaper, P.M.; Clay-Farrace, L.; Fiegler, H.; Carr, P.; Von Zglinicki, T.; Saretzki, G.; Carter, N.P.; Jackson, S.P. A DNA damage checkpoint response in telomere-initiated senescence. Nature 2003, 426, 194–198. [Google Scholar] [CrossRef] [PubMed]
- Chou, J.P.; Effros, R.B. T Cell Replicative Senescence in Human Aging. Curr. Pharm. Des. 2013, 19, 1680–1698. [Google Scholar] [CrossRef]
- Briceno, O.; Lissina, A.; Wanke, K.; Afonso, G.; Von Braun, A.; Ragon, K.; Miquel, T.; Gostick, E.; Papagno, L.; Stiasny, K.; et al. Reduced naïve CD8+ T-cell priming efficacy in elderly adults. Aging Cell 2016, 15, 14–21. [Google Scholar] [CrossRef] [PubMed]
- Jiang, J.; Fisher, E.M.; Murasko, D.M. CD8 T cell responses to influenza virus infection in aged mice. Ageing Res. Rev. 2011, 10, 422–427. [Google Scholar] [CrossRef] [Green Version]
- Brenchley, J.M.; Karandikar, N.J.; Betts, M.R.; Ambrozak, D.R.; Hill, B.J.; Crotty, L.E.; Casazza, J.P.; Kuruppu, J.; Migueles, S.A.; Connors, M.; et al. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood 2003, 101, 2711–2720. [Google Scholar] [CrossRef]
- Wang, Z.; Aguilar, E.G.; Luna, J.I.; Dunai, C.; Khuat, L.T.; Le, C.T.; Mirsoian, A.; Minnar, C.M.; Stoffel, K.M.; Sturgill, I.R.; et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat. Med. 2019, 25, 141–151. [Google Scholar] [CrossRef] [PubMed]
- Valenzuela, H. Divergent Telomerase and CD28 Expression Patterns in Human CD4 and CD8 T Cells Following Repeated Encounters with the Same Antigenic Stimulus. Clin. Immunol. 2002, 105, 117–125. [Google Scholar] [CrossRef]
- Renkema, K.R.; Li, G.; Wu, A.; Smithey, M.J.; Nikolich-Zugich, J. Two Separate Defects Affecting True Naive or Virtual Memory T Cell Precursors Combine To Reduce Naive T Cell Responses with Aging. J. Immunol. 2014, 192, 151–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Decman, V.; Laidlaw, B.J.; DiMenna, L.J.; Abdulla, S.; Mozdzanowska, K.; Erikson, J.; Ertl, H.C.J.; Wherry, E.J. Cell-Intrinsic Defects in the Proliferative Response of Antiviral Memory CD8 T Cells in Aged Mice upon Secondary Infection. J. Immunol. 2010, 184, 5151–5159. [Google Scholar] [CrossRef]
- Quinn, K.M.; Fox, A.; Harland, K.L.; Russ, B.E.; Li, J.; Nguyen, T.H.O.; Loh, L.; Olshanksy, M.; Naeem, H.; Tsyganov, K.; et al. Age-Related Decline in Primary CD8+ T Cell Responses Is Associated with the Development of Senescence in Virtual Memory CD8+ T Cells. Cell Rep. 2018, 23, 3512–3524. [Google Scholar] [CrossRef]
- Kugel, C.H., III; Douglass, S.M.; Webster, M.R.; Kaur, A.; Liu, Q.; Yin, X.; Weiss, S.A.; Darvishian, F.; Al-Rohil, R.N.; Ndoye, A.; et al. Age Correlates with Response to Anti-PD1, Reflecting Age-Related Differences in Intratumoral Effector and Regulatory T-Cell Populations. Clin. Cancer Res. 2018, 24, 5347–5356. [Google Scholar] [CrossRef] [Green Version]
- Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
- Chiarion-Sileni, V.; Pigozzo, J.; Ascierto, P.; Grimaldi, A.M.; Maio, M.; Di Guardo, L.; Marchetti, P.; de Rosa, F.; Nuzzo, C.; Testori, A.; et al. Efficacy and safety of ipilimumab in elderly patients with pretreated advanced melanoma treated at Italian centres through the expanded access programme. J. Exp. Clin. Cancer Res. 2014, 33, 30. [Google Scholar] [CrossRef]
- Motzer, R.J.; Escudier, B.; George, S.; Hammers, H.J.; Srinivas, S.; Tykodi, S.S.; Sosman, J.A.; Plimack, E.R.; Procopio, G.; McDermott, D.F.; et al. Nivolumab versus everolimus in patients with advanced renal cell carcinoma: Updated results with long-term follow-up of the randomized, open-label, phase 3 CheckMate 025 trial. Cancer 2020, 126, 4156–4167. [Google Scholar] [CrossRef]
- Daste, A.; Domblides, C.; Gross-Goupil, M.; Chakiba, C.; Quivy, A.; Cochin, V.; De Mones, E.; Larmonier, N.; Soubeyran, P.; Ravaud, A. Immune checkpoint inhibitors and elderly people: A review. Eur. J. Cancer 2017, 82, 155–166. [Google Scholar] [CrossRef]
- McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [Green Version]
- Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44. [Google Scholar] [CrossRef] [Green Version]
- Castro, A.; Pyke, R.M.; Zhang, X.; Thompson, W.K.; Day, C.-P.; Alexandrov, L.B.; Zanetti, M.; Carter, H. Strength of immune selection in tumors varies with sex and age. Nat. Commun. 2020, 11, 1–9. [Google Scholar] [CrossRef]
- Moreira, A.; Gross, S.; Kirchberger, M.C.; Erdmann, M.; Schuler, G.; Heinzerling, L.; Groß, S. Senescence markers: Predictive for response to checkpoint inhibitors. Int. J. Cancer 2019, 144, 1147–1150. [Google Scholar] [CrossRef]
- Cherkassky, L.; Morello, A.; Villena-Vargas, J.; Feng, Y.; Dimitrov, D.S.; Jones, D.R.; Sadelain, M.; Adusumilli, P.S. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Investig. 2016, 126, 3130–3144. [Google Scholar] [CrossRef] [Green Version]
- Kasakovski, D.; Xu, L.; Li, Y. T cell senescence and CAR-T cell exhaustion in hematological malignancies. J. Hematol. Oncol. 2018, 11, 91. [Google Scholar] [CrossRef]
- Das, S.; Johnson, D.B. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 306–311. [Google Scholar] [CrossRef]
- Battram, A.M.; Bachiller, M.; Martin-Antonio, B. Senescence in the Development and Response to Cancer with Immunotherapy: A Double-Edged Sword. Int. J. Mol. Sci. 2020, 21, 4346. [Google Scholar] [CrossRef]
- Ovadya, Y.; Krizhanovsky, V. Strategies targeting cellular senescence. J. Clin. Investig. 2018, 128, 1247–1254. [Google Scholar] [CrossRef]
- Patnaik, A.; Haluska, P.; Tolcher, A.W.; Erlichman, C.; Papadopoulos, K.P.; Lensing, J.L.; Beeram, M.; Molina, J.R.; Rasco, D.; Arcos, R.R.; et al. A First-in-Human Phase I Study of the Oral p38 MAPK Inhibitor, Ralimetinib (LY2228820 Dimesylate), in Patients with Advanced Cancer. Clin. Cancer Res. 2015, 22, 1095–1102. [Google Scholar] [CrossRef] [Green Version]
- Ebert, P.J.; Cheung, J.; Yang, Y.; McNamara, E.; Hong, R.; Moskalenko, M.; Gould, S.E.; Maecker, H.; Irving, B.A.; Kim, J.M.; et al. MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity 2016, 44, 609–621. [Google Scholar] [CrossRef] [Green Version]
- Loi, S.; Dushyanthen, S.; Beavis, P.A.; Salgado, R.; Denkert, C.; Savas, P.; Combs, S.; Rimm, D.L.; Giltnane, J.M.; Estrada, M.V.; et al. RAS/MAPK Activation Is Associated with Reduced Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Therapeutic Cooperation Between MEK and PD-1/PD-L1 Immune Checkpoint Inhibitors. Clin. Cancer Res. 2015, 22, 1499–1509. [Google Scholar] [CrossRef] [Green Version]
- Chang, J.; Wang, Y.; Shao, L.; Laberge, R.-M.; Demaria, M.; Campisi, J.; Janakiraman, K.; Sharpless, N.E.; Ding, S.; Feng, W.; et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 2016, 22, 78–83. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015, 14, 644–658. [Google Scholar] [CrossRef]
- Fuhrmann-Stroissnigg, H.; Ling, Y.Y.; Zhao, J.; McGowan, S.J.; Zhu, Y.; Brooks, R.W.; Grassi, D.; Gregg, S.Q.; Stripay, J.L.; Dorronsoro, A.; et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat. Commun. 2017, 8, 422. [Google Scholar] [CrossRef]
- Deschênes-Simard, X.; Gaumont-Leclerc, M.-F.; Bourdeau, V.; Lessard, F.; Moiseeva, O.; Forest, V.; Igelmann, S.; Mallette, F.A.; Saba-El-Leil, M.K.; Meloche, S.; et al. Tumor suppressor activity of the ERK/MAPK pathway by promoting selective protein degradation. Genes Dev. 2013, 27, 900–915. [Google Scholar] [CrossRef] [Green Version]
- Astle, M.V.; Hannan, K.M.; Ng, P.Y.; Lee, R.S.; George, A.J.; Hsu, A.K.; Haupt, Y.; Hannan, R.D.; Pearson, R.B. AKT induces senescence in human cells via mTORC1 and p53 in the absence of DNA damage: Implications for targeting mTOR during malignancy. Oncogene 2011, 31, 1949–1962. [Google Scholar] [CrossRef] [Green Version]
- Dietsch, G.N.; Randall, T.D.; Gottardo, R.; Northfelt, D.W.; Ramanathan, R.K.; Cohen, P.A.; Manjarrez, K.L.; Newkirk, M.; Bryan, J.K.; Hershberg, R.M. Late Stage Cancer Patients Remain Highly Responsive to Immune Activation by the Selective TLR8 Agonist Motolimod (VTX-2337). Clin. Cancer Res. 2015, 21, 5445–5452. [Google Scholar] [CrossRef] [Green Version]
- Chow, L.Q.M.; Morishima, C.; Eaton, K.D.; Baik, C.S.; Goulart, B.H.; Anderson, L.N.; Manjarrez, K.L.; Dietsch, G.N.; Bryan, J.K.; Hershberg, R.M.; et al. Phase Ib Trial of the Toll-like Receptor 8 Agonist, Motolimod (VTX-2337), Combined with Cetuximab in Patients with Recurrent or Metastatic SCCHN. Clin. Cancer Res. 2017, 23, 2442–2450. [Google Scholar] [CrossRef] [Green Version]
Senescent T Cells | Exhausted T Cells | |
---|---|---|
Stimulus | Repetitive Ag Stimulation, Stress | Continuous Ag Stimulation |
Cytokine Secretion | ↑ IFN--γ, IL-6, IL-8, IL-10, TNF, TGF-β | ↓ IFN-γ, IL-2, TNF |
Surface Markers | ↑ CD57, Tim-3, TGIT, CD45RA, KLRG1, ↓ CD28, CD27 | ↑ PD-1, LAG-3, CD 160, 2B4, CTLA-4, Tim-3, TGIT |
Metabolism | ↑ Glycolysis, ↓ Mitochondrial Biogenesis | ↓ Glycolysis, ↓ Mitochondrial Biogenesis |
Transcriptional | T-bet | Eomes, NFAT, TOX, T-bet, Nr4a |
Effector Functions | ↓ Granzyme B, ↓Perforin | ↓ Granzyme B |
Phenotypic Characteristic | ↓ Proliferative Capacity, ↑ DNA Damage Molecules, ↑ SA-β-gal activity | ↓ Proliferative Capacity, Cell Cycle Arrest |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Liu, W.; Stachura, P.; Xu, H.C.; Bhatia, S.; Borkhardt, A.; Lang, P.A.; Pandyra, A.A. Senescent Tumor CD8+ T Cells: Mechanisms of Induction and Challenges to Immunotherapy. Cancers 2020, 12, 2828. https://doi.org/10.3390/cancers12102828
Liu W, Stachura P, Xu HC, Bhatia S, Borkhardt A, Lang PA, Pandyra AA. Senescent Tumor CD8+ T Cells: Mechanisms of Induction and Challenges to Immunotherapy. Cancers. 2020; 12(10):2828. https://doi.org/10.3390/cancers12102828
Chicago/Turabian StyleLiu, Wei, Paweł Stachura, Haifeng C. Xu, Sanil Bhatia, Arndt Borkhardt, Philipp A. Lang, and Aleksandra A. Pandyra. 2020. "Senescent Tumor CD8+ T Cells: Mechanisms of Induction and Challenges to Immunotherapy" Cancers 12, no. 10: 2828. https://doi.org/10.3390/cancers12102828
APA StyleLiu, W., Stachura, P., Xu, H. C., Bhatia, S., Borkhardt, A., Lang, P. A., & Pandyra, A. A. (2020). Senescent Tumor CD8+ T Cells: Mechanisms of Induction and Challenges to Immunotherapy. Cancers, 12(10), 2828. https://doi.org/10.3390/cancers12102828