Head and Neck Cancer Immunotherapy: Molecular Biological Aspects of Preclinical and Clinical Research
Abstract
:Simple Summary
Abstract
1. Introduction
2. Adoptive T-Cell Transfer
3. Molecular Biological Aspect of T-Cell Therapy
4. Role of T-Cell Therapy in Head and Neck Cancer
5. Limitations of Adoptive T-Cell Transfer
6. Molecular Biological Mechanisms of CTLA4 and PD1/PD-L1
7. Immune Checkpoint Therapy
8. Role of Immune Checkpoint Therapy in Head and Neck Cancer
9. Limitations of Immune Checkpoint Therapy
10. Interleukin Biology
11. Interleukin Therapy
12. Limitations of Interleukin Therapy
13. Cancer Vaccines
14. Role of Cancer Vaccines in Head and Neck Cancer
15. Limitations of Cancer Vaccines
16. Preclinical Models for Research: A Special Mention
17. Conclusions
Author Contributions
Funding
Acknowledgments
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, 20, 651–668. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tong, J.C.; Tan, T.W.; Ranganathan, S. Methods and protocols for prediction of immunogenic epitopes. Brief. Bioinform. 2007, 8, 96–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teng, M.W.; Swann, J.B.; Koebel, C.M.; Schreiber, R.D.; Smyth, M.J. Immune-mediated dormancy: An equilibrium with cancer. J. Leukoc. Biol. 2008, 84, 988–993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bindea, G.; Mlecnik, B.; Fridman, W.H.; Pagès, F.; Galon, J. Natural immunity to cancer in humans. Curr. Opin. Immunol. 2010, 22, 215–222. [Google Scholar] [CrossRef]
- Pagès, F.; Galon, J.; Dieu-Nosjean, M.C.; Tartour, E.; Sautès-Fridman, C.; Fridman, W.H. Immune infiltration in human tumors: A prognostic factor that should not be ignored. Oncogene 2010, 29, 1093–1102. [Google Scholar] [CrossRef] [Green Version]
- Southam, C.M.; Brunschwig, A.; Levin, A.G.; Dizon, Q.S. Effect of leukocytes on transplantability of human cancer. Cancer 1966, 19, 1743–1753. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Packard, B.S.; Aebersold, P.M.; Solomon, D.; Topalian, S.L.; Toy, S.T.; Simon, P.; Lotze, M.T.; Yang, J.C.; Seipp, C.A.; et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N. Engl. J. Med. 1988, 319, 1676–1680. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Yannelli, J.R.; Yang, J.C.; Topalian, S.L.; Schwartzentruber, D.J.; Weber, J.S.; Parkinson, D.R.; Seipp, C.A.; Einhorn, J.H.; White, D.E. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J. Natl. Cancer Inst. 1994, 86, 1159–1166. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef]
- Zacharakis, N.; Chinnasamy, H.; Black, M.; Xu, H.; Lu, Y.C.; Zheng, Z.; Pasetto, A.; Langhan, M.; Shelton, T.; Prickett, T.; et al. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat. Med. 2018, 24, 724–730. [Google Scholar] [CrossRef] [PubMed]
- Palmer, D.C.; Guittard, G.C.; Franco, Z.; Crompton, J.G.; Eil, R.L.; Patel, S.J.; Ji, Y.; Van Panhuys, N.; Klebanoff, C.A.; Sukumar, M.; et al. Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance. J. Exp. Med. 2015, 212, 2095–2113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perica, K.; Varela, J.C.; Oelke, M.; Schneck, J. Adoptive T cell immunotherapy for cancer. Rambam Maimonides Med. J. 2015, 6, e0004. [Google Scholar] [CrossRef] [PubMed]
- Garrido, F.; Aptsiauri, N.; Doorduijn, E.M.; Garcia Lora, A.M.; van Hall, T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr. Opin. Immunol. 2016, 39, 44–51. [Google Scholar] [CrossRef] [Green Version]
- Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
- Sengsayadeth, S.; Savani, B.N.; Oluwole, O.; Dholaria, B. Overview of approved CAR-T therapies, ongoing clinical trials, and its impact on clinical practice. EJHaem 2021, 3 (Suppl. S1), 6–10. [Google Scholar] [CrossRef] [PubMed]
- Mohanty, R.; Chowdhury, C.R.; Arega, S.; Sen, P.; Ganguly, P.; Ganguly, N. CAR T cell therapy: A new era for cancer treatment (Review). Oncol. Rep. 2019, 42, 2183–2195. [Google Scholar] [CrossRef]
- Ghorashian, S.; Jacoby, E.; De Moerloose, B.; Rives, S.; Bonney, D.; Shenton, G.; Bader, P.; Bodmer, N.; Quintana, A.M.; Herrero, B.; et al. Tisagenlecleucel therapy for relapsed or refractory B-cell acute lymphoblastic leukaemia in infants and children younger than 3 years of age at screening: An international, multicentre, retrospective cohort study. Lancet Haematol. 2022, 9, e766–e775. [Google Scholar] [CrossRef]
- Locke, F.L.; Miklos, D.B.; Jacobson, C.A.; Perales, M.A.; Kersten, M.J.; Oluwole, O.O.; Ghobadi, A.; Rapoport, A.P.; McGuirk, J.; Pagel, J.M.; et al. All ZUMA-7 Investigators and Contributing Kite Members. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. N. Engl. J. Med. 2022, 386, 640–654. [Google Scholar] [CrossRef]
- Khurana, A.; Dalland, J.C.; Young, J.R.; Inwards, D.J.; Paludo, J. Brexucabtagene autoleucel therapy induces complete remission in a primary refractory blastoid mantle cell lymphoma with neurolymphomatosis. Am. J. Hematol. 2021, 96, E298–E301. [Google Scholar] [CrossRef]
- Kamdar, M.; Solomon, S.R.; Arnason, J.; Johnston, P.B.; Glass, B.; Bachanova, V.; Ibrahimi, S.; Mielke, S.; Mutsaers, P.; Hernandez-Ilizaliturri, F.; et al. Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): Results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet 2022, 399, 2294–2308. [Google Scholar] [PubMed]
- Munshi, N.C.; Anderson, L.D., Jr.; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A.; et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384, 705–716. [Google Scholar] [CrossRef] [PubMed]
- Berdeja, J.G.; Madduri, D.; Usmani, S.Z.; Jakubowiak, A.; Agha, M.; Cohen, A.D.; Stewart, A.K.; Hari, P.; Htut, M.; Lesokhin, A.; et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): A phase 1b/2 open-label study. Lancet 2021, 398, 314–324. [Google Scholar] [CrossRef]
- Davila, M.L.; Sadelain, M. Biology and clinical application of CAR T cells for B cell malignancies. Int. J. Hematol. 2016, 104, 6–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guedan, S.; Chen, X.; Madar, A.; Carpenito, C.; McGettigan, S.E.; Frigault, M.J.; Lee, J.; Posey, A.D., Jr.; Scholler, J.; Scholler, N.; et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 2014, 124, 1070–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, D.G.; Powell, D.J. Pro-survival signaling via CD27 costimulation drives effective CAR T-cell therapy. Oncoimmunology 2012, 1, 547–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mata, M.; Gerken, C.; Nguyen, P.; Krenciute, G.; Spencer, D.M.; Gottschalk, S. Inducible Activation of MyD88 and CD40 in CAR T Cells Results in Controllable and Potent Antitumor Activity in Preclinical Solid Tumor Models. Cancer Discov. 2017, 7, 1306–1319. [Google Scholar] [CrossRef] [Green Version]
- Hombach, A.A.; Heiders, J.; Foppe, M.; Chmielewski, M.; Abken, H. OX40 costimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirected CD4(+) T cells. Oncoimmunology 2012, 1, 458–466. [Google Scholar] [CrossRef] [Green Version]
- Levine, B.L.; Miskin, J.; Wonnacott, K.; Keir, C. Global Manufacturing of CAR T Cell Therapy. Mol. Ther. Methods Clin. Dev. 2016, 4, 92–101. [Google Scholar] [CrossRef] [Green Version]
- Norberg, S.M.; Hinrichs, C.S. Advances in Adoptive Cell Therapy for Head and Neck Cancer. Otolaryngol. Clin. N. Am. 2021, 54, 761–768. [Google Scholar] [CrossRef]
- Mei, Z.; Zhang, K.; Lam, A.K.; Huang, J.; Qiu, F.; Qiao, B.; Zhang, Y. MUC1 as a target for CAR-T therapy in head and neck squamous cell carinoma. Cancer Med. 2020, 9, 640–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, Y.P.; Jin, L.; Bennett, K.B.; Wang, D.; Fredenburg, K.M.; Tseng, J.E.; Chang, L.J.; Huang, J.; Chan, E.K.L. CD70 as a target for chimeric antigen receptor T cells in head and neck squamous cell carcinoma. Oral Oncol. 2018, 78, 145–150. [Google Scholar] [CrossRef]
- Doran, S.L.; Stevanović, S.; Adhikary, S.; Gartner, J.J.; Jia, L.; Kwong, M.L.M.; Faquin, W.C.; Hewitt, S.M.; Sherry, R.M.; Yang, J.C.; et al. T-Cell Receptor Gene Therapy for Human Papillomavirus-Associated Epithelial Cancers: A First-in-Human, Phase I/II Study. J. Clin. Oncol. 2019, 37, 2759–2768. [Google Scholar] [CrossRef] [PubMed]
- Marcinkowski, B.; Stevanović, S.; Helman, S.R.; Norberg, S.M.; Serna, C.; Jin, B.; Gkitsas, N.; Kadakia, T.; Warner, A.; Davis, J.L.; et al. Cancer targeting by TCR gene-engineered T cells directed against Kita-Kyushu Lung Cancer Antigen-1. J. Immunother. Cancer 2019, 7, 229. [Google Scholar] [CrossRef]
- Effern, M.; Glodde, N.; Braun, M.; Liebing, J.; Boll, H.N.; Yong, M.; Bawden, E.; Hinze, D.; van den Boorn-Konijnenberg, D.; Daoud, M.; et al. Adoptive T Cell Therapy Targeting Different Gene Products Reveals Diverse and Context-Dependent Immune Evasion in Melanoma. Immunity 2020, 53, 564–580.e9. [Google Scholar] [CrossRef] [PubMed]
- Hegde, M.; Mukherjee, M.; Grada, Z.; Pignata, A.; Landi, D.; Navai, S.A.; Wakefield, A.; Fousek, K.; Bielamowicz, K.; Chow, K.K.; et al. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J. Clin. Investig. 2016, 126, 3036–3052. [Google Scholar] [CrossRef] [Green Version]
- Hegde, M.; Corder, A.; Chow, K.K.; Mukherjee, M.; Ashoori, A.; Kew, Y.; Zhang, Y.J.; Baskin, D.S.; Merchant, F.A.; Brawley, V.S.; et al. Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma. Mol. Ther. 2013, 21, 2087–2101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steentoft, C.; Migliorini, D.; King, T.R.; Mandel, U.; June, C.H.; Posey, A.D., Jr. Glycan-directed CAR-T cells. Glycobiology 2018, 28, 656–669. [Google Scholar] [CrossRef] [Green Version]
- Whilding, L.M.; Halim, L.; Draper, B.; Parente-Pereira, A.C.; Zabinski, T.; Davies, D.M.; Maher, J. CAR T-Cells Targeting the Integrin αvβ6 and Co-Expressing the Chemokine Receptor CXCR2 Demonstrate Enhanced Homing and Efficacy against Several Solid Malignancies. Cancers 2019, 11, 674. [Google Scholar] [CrossRef] [Green Version]
- Frey, N.V.; Porter, D.L. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Hematol. Am. Soc. Hematol. Educ. Program. 2016, 2016, 567–572. [Google Scholar] [CrossRef]
- Yin, Y.; Boesteanu, A.C.; Binder, Z.A.; Xu, C.; Reid, R.A.; Rodriguez, J.L.; Cook, D.R.; Thokala, R.; Blouch, K.; McGettigan-Croce, B.; et al. Checkpoint Blockade Reverses Anergy in IL-13Rα2 Humanized scFv-Based CAR T Cells to Treat Murine and Canine Gliomas. Mol. Ther. Oncolytics 2018, 11, 20–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dariavach, P.; Mattéi, M.G.; Golstein, P.; Lefranc, M.P. Human Ig superfamily CTLA-4 gene: Chromosomal localization and identity of protein sequence between murine and human CTLA-4 cytoplasmic domains. Eur. J. Immunol. 1988, 18, 1901–1905. [Google Scholar] [CrossRef] [PubMed]
- Lo, B.; Zhang, K.; Lu, W.; Zheng, L.; Zhang, Q.; Kanellopoulou, C.; Zhang, Y.; Liu, Z.; Fritz, J.M.; Marsh, R.; et al. AUTOIMMUNE DISEASE. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 2015, 349, 436–440. [Google Scholar] [CrossRef] [Green Version]
- Tai, X.; Van Laethem, F.; Sharpe, A.H.; Singer, A. Induction of autoimmune disease in CTLA-4-/- mice depends on a specific CD28 motif that is required for in vivo costimulation. Proc. Natl. Acad. Sci. USA 2007, 104, 13756–13761. [Google Scholar] [CrossRef] [Green Version]
- Chuang, E.; Lee, K.M.; Robbins, M.D.; Duerr, J.M.; Alegre, M.L.; Hambor, J.E.; Neveu, M.J.; Bluestone, J.A.; Thompson, C.B. Regulation of cytotoxic T lymphocyte-associated molecule-4 by Src kinases. J. Immunol. 1999, 162, 1270–1277. [Google Scholar] [CrossRef]
- Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992, 11, 3887–3895. [Google Scholar] [CrossRef]
- Francisco, L.M.; Sage, P.T.; Sharpe, A.H. The PD-1 pathway in tolerance and autoimmunity. Immunol. Rev. 2010, 236, 219–242. [Google Scholar] [CrossRef]
- Hui, E.; Cheung, J.; Zhu, J.; Su, X.; Taylor, M.J.; Wallweber, H.A.; Sasmal, D.K.; Huang, J.; Kim, J.M.; Mellman, I.; et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355, 1428–1433. [Google Scholar] [CrossRef]
- Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell. Biol. 2005, 25, 9543–9553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McDermott, D.; Haanen, J.; Chen, T.T.; Lorigan, P.; O’Day, S.; MDX010-20 Investigators. Efficacy and safety of ipilimumab in metastatic melanoma patients surviving more than 2 years following treatment in a phase III trial (MDX010-20). Ann. Oncol. 2013, 24, 2694–2698. [Google Scholar] [CrossRef]
- Postow, M.A.; Chesney, J.; Pavlick, A.C.; Robert, C.; Grossmann, K.; McDermott, D.; Linette, G.P.; Meyer, N.; Giguere, J.K.; Agarwala, S.S.; et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N. Engl. J. Med. 2015, 372, 2006–2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weber, J.S.; D’Angelo, S.P.; Minor, D.; Hodi, F.S.; Gutzmer, R.; Neyns, B.; Hoeller, C.; Khushalani, N.I.; Miller, W.H., Jr.; Lao, C.D.; et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015, 16, 375–384. [Google Scholar] [CrossRef] [PubMed]
- Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 2015, 372, 311–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, P.; Retz, M.; Siefker-Radtke, A.; Baron, A.; Necchi, A.; Bedke, J.; Plimack, E.R.; Vaena, D.; Grimm, M.-O.; Galsky, M.D.; et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): Amulticentre, single-arm, phase 2 trial. Lancet Oncol. 2017, 18, 312–322. [Google Scholar] [CrossRef]
- Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A.; et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef]
- El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.Y.; Choo, S.P.; Trojan, J.; Welling, T.H., 3rd; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef]
- Robert, C.; Ribas, A.; Wolchok, J.D.; Hodi, F.S.; Hamid, O.; Kefford, R.; Weber, J.S.; Joshua, A.M.; Hwu, W.J.; Gangadhar, T.C.; et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: A randomised dose-comparison cohort of a phase 1 trial. Lancet 2014, 384, 1109–1117. [Google Scholar] [CrossRef]
- Migden, M.R.; Rischin, D.; Schmults, C.D.; Guminski, A.; Hauschild, A.; Lewis, K.D.; Chung, C.H.; Hernandez-Aya, L.; Lim, A.M.; Chang, A.L.S.; et al. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N. Engl. J. Med. 2018, 379, 341–351. [Google Scholar] [CrossRef] [Green Version]
- Kaufman, H.L.; Russell, J.; Hamid, O.; Bhatia, S.; Terheyden, P.; D’Angelo, S.P.; Shih, K.C.; Lebbé, C.; Linette, G.P.; Milella, M.; et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: A multicentre, single-group, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 1374–1385. [Google Scholar] [CrossRef]
- Massard, C.; Gordon, M.S.; Sharma, S.; Rafii, S.; Wainberg, Z.A.; Luke, J.; Curiel, T.J.; Colon-Otero, G.; Hamid, O.; Sanborn, R.E.; et al. Safety and Efficacy of Durvalumab (MEDI4736), an Anti-Programmed Cell Death Ligand-1 Immune Checkpoint Inhibitor, in Patients With Advanced Urothelial Bladder Cancer. J. Clin. Oncol. 2016, 34, 3119–3125. [Google Scholar] [CrossRef] [Green Version]
- Necchi, A.; Joseph, R.W.; Loriot, Y.; Hoffman-Censits, J.; Perez-Gracia, J.L.; Petrylak, D.P.; Derleth, C.L.; Tayama, D.; Zhu, Q.; Ding, B.; et al. Atezolizumab in platinum-treated locally advanced or metastatic urothelial carcinoma: Post-progression outcomes from the phase II IMvigor210 study. Ann. Oncol. 2017, 28, 3044–3050. [Google Scholar] [CrossRef]
- Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef] [PubMed]
- de Sousa, L.G.; Ferrarotto, R. Pembrolizumab in the first-line treatment of advanced head and neck cancer. Expert Rev. Anticancer Ther. 2021, 21, 1321–1331. [Google Scholar] [CrossRef] [PubMed]
- Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G.; Psyrri, A.; Basté, N.; Neupane, P.; Abel, E.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
- Ferris, R.L.; Moskovitz, J.; Kunning, S.; Ruffin, A.T.; Reeder, C.; Ohr, J.; Gooding, W.E.; Kim, S.; Karlovits, B.J.; Vignali, D.A.A.; et al. Phase I Trial of Cetuximab, Radiotherapy, and Ipilimumab in Locally Advanced Head and Neck Cancer. Clin. Cancer Res. 2022, 28, 1335–1344. [Google Scholar] [CrossRef] [PubMed]
- Fares, C.M.; Van Allen, E.M.; Drake, C.G.; Allison, J.P.; Hu-Lieskovan, S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 147–164. [Google Scholar] [CrossRef]
- Dobosz, P.; Stępień, M.; Golke, A.; Dzieciątkowski, T. Challenges of the Immunotherapy: Perspectives and Limitations of the Immune Checkpoint Inhibitor Treatment. Int. J. Mol. Sci. 2022, 23, 2847. [Google Scholar] [CrossRef]
- Park, J.A.; Cheung, N.V. Limitations and opportunities for immune checkpoint inhibitors in pediatric malignancies. Cancer Treat. Rev. 2017, 58, 22–33. [Google Scholar] [CrossRef]
- Briukhovetska, D.; Dörr, J.; Endres, S.; Libby, P.; Dinarello, C.A.; Kobold, S. Interleukins in cancer: From biology to therapy. Nat. Rev. Cancer 2021, 21, 481–499. [Google Scholar] [CrossRef]
- Lamkanfi, M.; Dixit, V.M. Mechanisms and functions of inflammasomes. Cell 2014, 157, 1013–1022. [Google Scholar] [CrossRef] [Green Version]
- Tu, S.; Bhagat, G.; Cui, G.; Takaishi, S.; Kurt-Jones, E.A.; Rickman, B.; Betz, K.S.; Penz-Oesterreicher, M.; Bjorkdahl, O.; Fox, J.G.; et al. Overexpression of interleukin-1beta induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice. Cancer Cell 2008, 14, 408–419. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Barajon, I.; Garlanda, C. IL-1 and IL-1 regulatory pathways in cancer progression and therapy. Immunol. Rev. 2018, 281, 57–61. [Google Scholar] [CrossRef]
- Jiang, T.; Zhou, C.; Ren, S. Role of IL-2 in cancer immunotherapy. Oncoimmunology 2016, 5, e1163462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mangi, M.H.; Newland, A.C. Interleukin-3 in hematology and oncology: Current state of knowledge and future directions. Cytokines Cell. Mol. Ther. 1999, 5, 87–95. [Google Scholar] [PubMed]
- Nappo, G.; Handle, F.; Santer, F.R.; McNeill, R.V.; Seed, R.I.; Collins, A.T.; Morrone, G.; Culig, Z.; Maitland, N.J.; Erb, H.H.H. The immunosuppressive cytokine interleukin-4 increases the clonogenic potential of prostate stem-like cells by activation of STAT6 signalling. Oncogenesis 2017, 6, e342. [Google Scholar] [CrossRef] [Green Version]
- Zaynagetdinov, R.; Sherrill, T.P.; Gleaves, L.A.; McLoed, A.G.; Saxon, J.A.; Habermann, A.C.; Connelly, L.; Dulek, D.; Peebles, R.S., Jr.; Fingleton, B. Interleukin-5 facilitates lung metastasis by modulating the immune microenvironment. Cancer Res. 2015, 75, 1624–1634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petes, C.; Mariani, M.K.; Yang, Y.; Grandvaux, N.; Gee, K. Interleukin (IL)-6 Inhibits IL-27- and IL-30-Mediated Inflammatory Responses in Human Monocytes. Front. Immunol. 2018, 9, 256. [Google Scholar] [CrossRef] [Green Version]
- Lin, J.; Zhu, Z.; Xiao, H.; Wakefield, M.R.; Ding, V.A.; Bai, Q.; Fang, Y. The role of IL-7 in Immunity and Cancer. Anticancer Res. 2017, 37, 963–967. [Google Scholar]
- Alfaro, C.; Sanmamed, M.F.; Rodríguez-Ruiz, M.E.; Teijeira, Á.; Oñate, C.; González, Á.; Ponz, M.; Schalper, K.A.; Pérez-Gracia, J.L.; Melero, I. Interleukin-8 in cancer pathogenesis, treatment and follow-up. Cancer Treat. Rev. 2017, 60, 24–31. [Google Scholar] [CrossRef]
- Hu, B.; Qiu-Lan, H.; Lei, R.E.; Shi, C.; Jiang, H.X.; Qin, S.Y. Interleukin-9 Promotes Pancreatic Cancer Cells Proliferation and Migration via the miR-200a/Beta-Catenin Axis. BioMed. Res. Int. 2017, 2017, 2831056. [Google Scholar] [CrossRef]
- Mannino, M.H.; Zhu, Z.; Xiao, H.; Bai, Q.; Wakefield, M.R.; Fang, Y. The paradoxical role of IL-10 in immunity and cancer. Cancer Lett. 2015, 367, 103–107. [Google Scholar] [CrossRef] [PubMed]
- Johnstone, C.N.; Chand, A.; Putoczki, T.L.; Ernst, M. Emerging roles for IL-11 signaling in cancer development and progression: Focus on breast cancer. Cytokine Growth Factor Rev. 2015, 26, 489–498. [Google Scholar] [CrossRef] [Green Version]
- Lu, X. Impact of IL-12 in Cancer. Curr. Cancer Drug Targets 2017, 17, 682–697. [Google Scholar] [CrossRef]
- Terabe, M.; Park, J.M.; Berzofsky, J.A. Role of IL-13 in regulation of anti-tumor immunity and tumor growth. Cancer Immunol. Immunother. 2004, 53, 79–85. [Google Scholar] [CrossRef] [PubMed]
- Ford, R.; Tamayo, A.; Martin, B.; Niu, K.; Claypool, K.; Cabanillas, F.; Ambrus, J., Jr. Identification of B-cell growth factors (interleukin-14; high molecular weight-B-cell growth factors) in effusion fluids from patients with aggressive B-cell lymphomas. Blood 1995, 86, 283–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, W.; Liu, N.; Yuan, Y.; Zhu, M.; Hu, X.; Hu, W.; Wang, S.; Wang, C.; Huang, B.; Xing, D. ALT-803 in the treatment of non-muscle-invasive bladder cancer: Preclinical and clinical evidence and translational potential. Front. Immunol. 2022, 13, 1040669. [Google Scholar] [CrossRef]
- Richmond, J.; Richmond, J.; Tuzova, M.; Cruikshank, W.; Center, D. Regulation of cellular processes by interleukin-16 in homeostasis and cancer. J. Cell. Physiol. 2014, 229, 139–147. [Google Scholar] [CrossRef]
- Yang, B.; Kang, H.; Fung, A.; Zhao, H.; Wang, T.; Ma, D. The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis. Mediat. Inflamm. 2014, 2014, 623759. [Google Scholar] [CrossRef] [PubMed]
- Alboni, S.; Montanari, C.; Benatti, C.; Sanchez-Alavez, M.; Rigillo, G.; Blom, J.M.; Brunello, N.; Conti, B.; Pariante, M.C.; Tascedda, F. Interleukin 18 activates MAPKs and STAT3 but not NF-κB in hippocampal HT-22 cells. Brain Behav. Immun. 2014, 40, 85–94. [Google Scholar] [CrossRef]
- Hsing, C.H.; Cheng, H.C.; Hsu, Y.H.; Chan, C.H.; Yeh, C.H.; Li, C.F.; Chang, M.S. Upregulated IL-19 in breast cancer promotes tumor progression and affects clinical outcome. Clin. Cancer Res. 2012, 18, 713–725. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.J.; Cho, S.C.; Lee, E.J.; Kim, S.; Lee, S.B.; Lim, J.H.; Choi, Y.H.; Kim, W.J.; Moon, S.K. Interleukin-20 promotes migration of bladder cancer cells through extracellular signal-regulated kinase (ERK)-mediated MMP-9 protein expression leading to nuclear factor (NF-κB) activation by inducing the up-regulation of p21(WAF1) protein expression. J. Biol. Chem. 2013, 288, 5539–5552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wan, C.K.; Andraski, A.B.; Spolski, R.; Li, P.; Kazemian, M.; Oh, J.; Samsel, L.; Swanson, P.A., 2nd; McGavern, D.B.; Sampaio, E.P.; et al. Opposing roles of STAT1 and STAT3 in IL-21 function in CD4+ T cells. Proc. Natl. Acad. Sci. USA 2015, 112, 9394–9399. [Google Scholar] [CrossRef] [Green Version]
- Madonna, S.; Scarponi, C.; Morelli, M.; Sestito, R.; Scognamiglio, P.L.; Marasco, D.; Albanesi, C. SOCS3 inhibits the pathological effects of IL-22 in non-melanoma skin tumor-derived keratinocytes. Oncotarget 2017, 8, 24652–24667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Revu, S.; Wu, J.; Henkel, M.; Rittenhouse, N.; Menk, A.; Delgoffe, G.M.; Poholek, A.C.; McGeachy, M.J. IL-23 and IL-1β Drive Human Th17 Cell Differentiation and Metabolic Reprogramming in Absence of CD28 Costimulation. Cell Rep. 2018, 22, 2642–2653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kreist, S.; Philippidou, D.; Margue, C.; Behrmann, I. IL-24: A classic cytokine and/or a potential cure for cancer? J. Cell. Mol. Med. 2008, 12, 2505–2510. [Google Scholar] [CrossRef]
- Dambacher, J.; Beigel, F.; Zitzmann, K.; De Toni, E.N.; Göke, B.; Diepolder, H.M.; Auernhammer, C.J.; Brand, S. The role of the novel Th17 cytokine IL-26 in intestinal inflammation. Gut 2009, 58, 1207–1217. [Google Scholar] [CrossRef] [Green Version]
- Fabbi, M.; Carbotti, G.; Ferrini, S. Dual Roles of IL-27 in Cancer Biology and Immunotherapy. Mediat. Inflamm. 2017, 2017, 3958069. [Google Scholar] [CrossRef] [Green Version]
- Wongthida, P.; Diaz, R.M.; Galivo, F.; Kottke, T.; Thompson, J.; Pulido, J.; Pavelko, K.; Pease, L.; Melcher, A.; Vile, R. Type III IFN interleukin-28 mediates the antitumor efficacy of oncolytic virus VSV in immune-competent mouse models of cancer. Cancer Res. 2010, 70, 4539–4549. [Google Scholar] [CrossRef] [Green Version]
- Airoldi, I.; Cocco, C.; Sorrentino, C.; Angelucci, D.; Di Meo, S.; Manzoli, L.; Esposito, S.; Ribatti, D.; Bertolotto, M.; Iezzi, L.; et al. Interleukin-30 Promotes Breast Cancer Growth and Progression. Cancer Res. 2016, 76, 6218–6229. [Google Scholar] [CrossRef] [Green Version]
- Ferretti, E.; Corcione, A.; Pistoia, V. The IL-31/IL-31 receptor axis: General features and role in tumor microenvironment. J. Leukoc. Biol. 2017, 102, 711–717. [Google Scholar] [CrossRef]
- Yan, H.; He, D.; Huang, X.; Zhang, E.; Chen, Q.; Xu, R.; Liu, X.; Zi, F.; Cai, Z. Role of interleukin-32 in cancer biology. Oncol. Lett. 2018, 16, 41–47. [Google Scholar] [CrossRef] [Green Version]
- Jovanovic, I.P.; Pejnovic, N.N.; Radosavljevic, G.D.; Arsenijevic, N.N.; Lukic, M.L. IL-33/ST2 axis in innate and acquired immunity to tumors. Oncoimmunology 2012, 1, 229–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baghdadi, M.; Endo, H.; Takano, A.; Ishikawa, K.; Kameda, Y.; Wada, H.; Miyagi, Y.; Yokose, T.; Ito, H.; Nakayama, H.; et al. High co-expression of IL-34 and M-CSF correlates with tumor progression and poor survival in lung cancers. Sci. Rep. 2018, 8, 418. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Liu, J.Q.; Liu, Z.; Shen, R.; Zhang, G.; Xu, J.; Basu, S.; Feng, Y.; Bai, X.F. Tumor-derived IL-35 promotes tumor growth by enhancing myeloid cell accumulation and angiogenesis. J. Immunol. 2013, 190, 2415–2423. [Google Scholar] [CrossRef] [Green Version]
- Müller, A.; Hennig, A.; Lorscheid, S.; Grondona, P.; Schulze-Osthoff, K.; Hailfinger, S.; Kramer, D. IκBζ is a key transcriptional regulator of IL-36-driven psoriasis-related gene expression in keratinocytes. Proc. Natl. Acad. Sci. USA 2018, 115, 10088–10093. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Quan, Y.; Yue, Y.; Heng, X.; Che, F. Interleukin-37: A crucial cytokine with multiple roles in disease and potentially clinical therapy. Oncol. Lett. 2018, 15, 4711–4719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takada, K.; Okamoto, T.; Tominaga, M.; Teraishi, K.; Akamine, T.; Takamori, S.; Katsura, M.; Toyokawa, G.; Shoji, F.; Okamoto, M.; et al. Clinical implications of the novel cytokine IL-38 expressed in lung adenocarcinoma: Possible association with PD-L1 expression. PLoS ONE 2017, 12, e0181598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ridker, P.M.; MacFadyen, J.G.; Thuren, T.; Everett, B.M.; Libby, P.; Glynn, R.J. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: Exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 2017, 390, 1833–1842. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, M.L.T.; Bui, K.C.; Scholta, T.; Xing, J.; Bhuria, V.; Sipos, B.; Wilkens, L.; Nguyen Linh, T.; Velavan, T.P.; Bozko, P.; et al. Targeting interleukin 6 signaling by monoclonal antibody siltuximab on cholangiocarcinoma. J. Gastroenterol. Hepatol. 2021, 36, 1334–1345. [Google Scholar] [CrossRef] [PubMed]
- Orlowski, R.Z.; Gercheva, L.; Williams, C.; Sutherland, H.; Robak, T.; Masszi, T.; Goranova-Marinova, V.; Dimopoulos, M.A.; Cavenagh, J.D.; Špička, I.; et al. A phase 2, randomized, double-blind, placebo-controlled study of siltuximab (anti-IL-6 mAb) and bortezomib versus bortezomib alone in patients with relapsed or refractory multiple myeloma. Am. J. Hematol. 2015, 90, 42–49. [Google Scholar] [CrossRef] [PubMed]
- Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef] [PubMed]
- Nagaoka, K.; Shirai, M.; Taniguchi, K.; Hosoi, A.; Sun, C.; Kobayashi, Y.; Maejima, K.; Fujita, M.; Nakagawa, H.; Nomura, S.; et al. Deep immunophenotyping at the single-cell level identifies a combination of anti-IL-17 and checkpoint blockade as an effective treatment in a preclinical model of data-guided personalized immunotherapy. J. Immunother. Cancer 2020, 8, e001358. [Google Scholar] [CrossRef] [PubMed]
- Prabhala, R.H.; Fulciniti, M.; Pelluru, D.; Rashid, N.; Nigroiu, A.; Nanjappa, P.; Pai, C.; Lee, S.; Prabhala, N.S.; Bandi, R.L.; et al. Targeting IL-17A in multiple myeloma: A potential novel therapeutic approach in myeloma. Leukemia 2016, 30, 379–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Calcinotto, A.; Spataro, C.; Zagato, E.; Di Mitri, D.; Gil, V.; Crespo, M.; De Bernardis, G.; Losa, M.; Mirenda, M.; Pasquini, E.; et al. IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature 2018, 559, 363–369. [Google Scholar] [CrossRef]
- Zhou, J.Z.; Jou, J.; Cohen, E. Vaccine Strategies for Human Papillomavirus-Associated Head and Neck Cancers. Cancers 2021, 14, 33. [Google Scholar] [CrossRef]
- Beyaert, S.; Machiels, J.P.; Schmitz, S. Vaccine-Based Immunotherapy for Head and Neck Cancers. Cancers 2021, 13, 6041. [Google Scholar] [CrossRef]
- Shibata, H.; Zhou, L.; Xu, N.; Egloff, A.M.; Uppaluri, R. Personalized cancer vaccination in head and neck cancer. Cancer Sci. 2021, 112, 978–988. [Google Scholar] [CrossRef]
- Bareham, B.; Georgakopoulos, N.; Matas-Céspedes, A.; Curran, M.; Saeb-Parsy, K. Modeling human tumor-immune environments in vivo for the preclinical assessment of immunotherapies. Cancer Immunol. Immunother. 2021, 70, 2737–2750. [Google Scholar] [CrossRef]
- Munnik, C.; Xaba, M.P.; Malindisa, S.T.; Russell, B.L.; Sooklal, S.A. Drosophila melanogaster: A platform for anticancer drug discovery and personalized therapies. Front. Genet. 2022, 13, 949241. [Google Scholar] [CrossRef]
- Hidalgo, M.; Amant, F.; Biankin, A.V.; Budinská, E.; Byrne, A.T.; Caldas, C.; Clarke, R.B.; de Jong, S.; Jonkers, J.; Mælandsmo, G.M.; et al. Patient-derived xenograft models: An emerging platform for translational cancer research. Cancer Discov. 2014, 4, 998–1013. [Google Scholar] [CrossRef] [PubMed]
- Rasmussen, J.H.; Lelkaitis, G.; Håkansson, K.; Vogelius, I.R.; Johannesen, H.H.; Fischer, B.M.; Bentzen, S.M.; Specht, L.; Kristensen, C.A.; von Buchwald, C.; et al. Intratumor heterogeneity of PD-L1 expression in head and neck squamous cell carcinoma. Br. J. Cancer 2019, 120, 1003–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karpathiou, G.; Vincent, M.; Dumollard, J.M.; Mobarki, M.; Péoc’h, M. PD-L1 expression in head and neck cancer tissue specimens decreases with time. Pathol. Res. Pract. 2022, 237, 154042. [Google Scholar] [CrossRef] [PubMed]
Type of Cancer | Immune Checkpoint Blocker |
---|---|
Squamous cell head and neck carcinoma | Nivolumab or pembrolizumab |
Malignant melanoma | Ipilimumab, nivolumab, pembrolizumab |
Merkel cell carcinoma | Avelumab, pembrolizumab |
Hepatocellular carcinoma | Nivolumab, pembrolizumab |
Cutaneous squamous cell carcinoma | Cemiplimab |
Advanced renal carcinoma | Nivolumab or ipilimumab |
Colorectal cancer | Nivolumab or ipilimumab |
Cervical cancer | Pembrolizumab |
Small cell lung cancer | Atezolizumab, nivolumab |
Non-small cell lung cancer | Durvalumab, pembrolizumab, atezolizumab, nivolumab |
Triple-negative breast cancer | Atezolizumab |
Gastric carcinoma | Pembrolizumab |
Hodgkin lymphoma | Pembrolizumab |
Primary mediastinal large B-cell lymphoma | Pembrolizumab |
Metastatic urothelial cancer | Durvalumab, pembrolizumab, atezolizumab, nivolumab, avelumab |
Interleukin | Effect of Interleukins in Cancer | Reference |
---|---|---|
IL-1 | VEGF angiogenesis; MMP metastasis; COX-2, iNOS, PGE2, and IL-17 induction | [73] |
IL-2 | A growth factor for Teffector cells; promotes antitumour immunity | [74] |
IL-3 | Activation of c-MYC, c-Fos, and c-FMS; protumour effect; helps in anti-apoptosis, and proliferation; affects osteoclast/osteoblast formation; aids in survival of malignant haematopoietic cells | [75] |
IL-4 | Angiogenesis; increases clonogenic potential of stem cells, as well as AKT, p44/42 MAPK, NF-κB, and JAK/STAT6 pathways; immunosuppression | [76] |
IL-5 | Facilitates metastasis; overexpression indicates poor prognosis | [77] |
IL-6 | Signalling of cell survival, proliferation, metastasis, and angiogenesis, with profound effects on cancer | [78] |
IL-7 | Antiapoptotic and proliferation | [79] |
IL-8 | Assists in progression of cancer; affects gene expression; modulates translation and post-translational modifications of proteins, affecting cytoskeletal organisation, tumour immune resistance, and angiogenesis; accentuates proliferation signals and attracts myeloid cells to provide growth factors | [80] |
IL-9 | Induces TH9 cells, STAT6, IRF4, and PU.1; regulates intestinal barrier function via regulation of claudin-2. | [81] |
IL-10 | Plays a role in the regulation of various subsets of CD4+ T cells; in combination with IL-4 and IL-2 helps in proliferation of CD8+ T cells; IL-10-treated DCs induce an anergic state in CD4+ and CD8+ T cells; IL-10/STAT3 mediates anti-inflammatory response and promotes survival of B cells, MHC class II suppression, with both tumour promotion and inhibitory roles | [82] |
IL-11 | Aids in the progression of cancer by influencing the IL-11/STAT3 axis | [83] |
IL-12 | Tumour suppression; increases antiangiogenesis and antiapoptotic factors; increases IL-2R and IFNγ. | [84] |
IL-13 | Evasion and metastasis; suppresses CTL responses against cancer; facilitates growth of cancer. | [85] |
IL-14 | B-cell proliferation | [86] |
IL-15 | Antitumour immunity | [87] |
IL-16 | Chemoattractant; growth factor | [88] |
IL-17 | Angiogenesis, metastasis, and lipocalin | [89] |
IL-18 | Modulates TH1 differentiation, pro-inflammatory activity, and NK-mediated cytotoxicity; upregulates IFN-γ, TNF-α, IL-1β, and IL-8; activates MAPK and STAT3 | [90] |
IL-19 | Induces JNK, ERK, Akt, NF-κB, STAT1, and STAT3; upregulates MMP2 and MMP9; promotes proliferation of oral squamous cell carcinoma | [91] |
IL-20 | Increases migration and proliferation of cancers through activation of bFGF, VEGF, STAT3, ERK, JNK, and Bcl-xL | [92] |
IL-21 | Activates the JAK/STAT, PI3K, and MAPK pathways; enhances cytotoxic activity of NK and NKT cells; induces DC apoptosis | [93] |
IL-22 | Upregulates proliferative pathways such as JAK/STAT, PI3K/Akt, NF-κB, MAPK, and mTOR | [94] |
IL-23 | IL-23 and TGFβ suppress metastasis in pancreatic cancer; IL-23 and IL-1β drive TH17 differentiation and promote growth and progression of oral squamous cell carcinoma | [95] |
IL-24 | Tumour suppressor; suppression of glial inflammatory response | [96] |
IL-26 | Proinflammatory and prosurvival activities by regulating the balance of STAT1 and STAT3 activity | [97] |
IL-27 | Anticancer effect; reduction in VEGF and a series of angiopoietins; inhibits Treg; downregulates MMP9; inhibits COX-2 and PGE2 | [98] |
IL-28 | IL-28A promotes upregulation of MHC I and suppression of Th 17 and Th 2 responses | [99] |
IL-30 | Binds to gp130 and blocks pathways affected by IL-6 and IL-27. Binding to gp130 activates STAT1/STAT3 and promotes cancer progression; promotes BRCA cell migration and invasiveness | [100] |
IL-31 | The IL-31/IL-31R axis recruits OSMR, activating MAPK and PI3K/Akt | [101] |
IL-32 | Overexpression in HNSCC induces TNFα, IL-6, and IL-1β, as well as macrophage inflammatory protein-2 (MIP-2) | [102] |
IL-33 | IL-33/ST2 activates TH2-polarized cells, leading to the induction of IL-4, IL-10, and IL-13; generates immature DCs, which induce Treg | [103] |
IL-34 | High coexpression of IL-34 and M-CSF correlates with poor prognosis in lung cancer patients; promotes tumour progression and metastasis via induction of angiogenesis and macrophage recruitment | [104] |
IL-35 | CD11b (+) Gr1(+) myeloid cells promote cancer; cell cycle arrest in G1 phase; apoptosis due to TNFα; IFNγ-mediated upregulation of Fas ligand with simultaneous downregulation of cyclin D1, survivin, and Bcl-2 | [105] |
IL-36 | IL-36α suppresses proliferation of cancer cells; exerts proinflammatory activity through MAPK and NF-κB | [106] |
IL-37 | Inhibits T-cell and DC activation; promotes IL-10 and IL-16; regulates STAT3; promotes Treg; inhibits Smad3/TGFβ; inhibits proliferation and invasion of cancer cells | [107] |
IL-38 | Inhibits IL-8, IL-17, and IL-22; overexpression is related to poor prognosis | [108] |
Treatment Option | Progress |
---|---|
Adoptive T-cell transfer |
|
Immune checkpoint therapy |
|
Interleukin therapy |
|
Cancer vaccines |
|
Personalized therapy |
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chakraborty, R.; Darido, C.; Liu, F.; Maselko, M.; Ranganathan, S. Head and Neck Cancer Immunotherapy: Molecular Biological Aspects of Preclinical and Clinical Research. Cancers 2023, 15, 852. https://doi.org/10.3390/cancers15030852
Chakraborty R, Darido C, Liu F, Maselko M, Ranganathan S. Head and Neck Cancer Immunotherapy: Molecular Biological Aspects of Preclinical and Clinical Research. Cancers. 2023; 15(3):852. https://doi.org/10.3390/cancers15030852
Chicago/Turabian StyleChakraborty, Rajdeep, Charbel Darido, Fei Liu, Maciej Maselko, and Shoba Ranganathan. 2023. "Head and Neck Cancer Immunotherapy: Molecular Biological Aspects of Preclinical and Clinical Research" Cancers 15, no. 3: 852. https://doi.org/10.3390/cancers15030852
APA StyleChakraborty, R., Darido, C., Liu, F., Maselko, M., & Ranganathan, S. (2023). Head and Neck Cancer Immunotherapy: Molecular Biological Aspects of Preclinical and Clinical Research. Cancers, 15(3), 852. https://doi.org/10.3390/cancers15030852