Vaccination against Cancer or Infectious Agents during Checkpoint Inhibitor Therapy
Abstract
:1. Introduction
2. Preventive Cancer Vaccines
3. Therapeutic Cancer Vaccines
4. Challenges of Therapeutic Cancer Vaccines
5. Immune Checkpoint Blockade and Combination with Therapeutic Cancer Vaccines
Potential Mechanisms to Increase Cancer Vaccine Efficacy
6. Safety and Efficacy of Cancer Vaccines in Combination with ICI
7. Vaccines against Infectious Agents in ICI-Treated Cancer Patients
7.1. Influenza Vaccines and ICIs
7.2. COVID-19 Vaccines and ICIs
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Zhivaki, D.; Borriello, F.; Chow, O.A.; Doran, B.; Fleming, I.; Theisen, D.J.; Pallis, P.; Shalek, A.K.; Sokol, C.L.; Zanoni, I.; et al. Inflammasomes within Hyperactive Murine Dendritic Cells Stimulate Long-Lived T Cell-Mediated Anti-tumor Immunity. Cell Rep. 2020, 33, 108381. [Google Scholar] [CrossRef]
- Wieland, A.; Patel, M.R.; Cardenas, M.A.; Eberhardt, C.S.; Hudson, W.H.; Obeng, R.C.; Griffith, C.C.; Wang, X.; Chen, Z.G.; Kissick, H.T.; et al. Defining HPV-specific B cell responses in patients with head and neck cancer. Nature 2020, 597, 274–278. [Google Scholar] [CrossRef]
- Kavanagh, K.; Pollock, K.G.; Cuschieri, K.; Palmer, T.; Cameron, R.L.; Watt, C.; Bhatia, R.; Moore, C.; Cubie, H.; Cruickshank, M.; et al. Changes in the prevalence of human papillomavirus following a national bivalent human papillomavirus vaccination programme in Scotland: A 7-year cross-sectional study. Lancet. Infect. Dis. 2017, 17, 1293–1302. [Google Scholar] [CrossRef][Green Version]
- Saslow, D.; Andrews, K.S.; Manassaram-Baptiste, D.; Smith, R.A.; Fontham, E.T.H. Human papillomavirus vaccination 2020 guideline update: American Cancer Society guideline adaptation. CA A Cancer J. Clin. 2020, 70, 274–280. [Google Scholar] [CrossRef] [PubMed]
- Drolet, M.; Benard, E.; Perez, N.; Brisson, M.; Group, H.P.V.V.I.S. Population-level impact and herd effects following the introduction of human papillomavirus vaccination programmes: Updated systematic review and meta-analysis. Lancet 2019, 394, 497–509. [Google Scholar] [CrossRef][Green Version]
- Herrero, R.; Quint, W.; Hildesheim, A.; Gonzalez, P.; Struijk, L.; Katki, H.A.; Porras, C.; Schiffman, M.; Rodriguez, A.C.; Solomon, D.; et al. Reduced prevalence of oral human papillomavirus (HPV) 4 years after bivalent HPV vaccination in a randomized clinical trial in Costa Rica. PLoS ONE 2013, 8, e68329. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Kreimer, A.R.; González, P.; Katki, H.A.; Porras, C.; Schiffman, M.; Rodriguez, A.C.; Solomon, D.; Jiménez, S.; Schiller, J.T.; Lowy, D.R.; et al. Efficacy of a bivalent HPV 16/18 vaccine against anal HPV 16/18 infection among young women: A nested analysis within the Costa Rica Vaccine Trial. Lancet. Oncol. 2011, 12, 862–870. [Google Scholar] [CrossRef][Green Version]
- Chang, M.H. Hepatitis B virus and cancer prevention. In Clinical Cancer Prevention; Springer: Berlin/Heidelberg, Germany, 2011; pp. 75–84. [Google Scholar] [CrossRef]
- Pyeon, D.; Vu, L.; Giacobbi, N.S.; Westrich, J.A. The antiviral immune forces awaken in the cancer wars. PLoS Pathog. 2020, 16, e1008814. [Google Scholar] [CrossRef]
- Nasti, T.H.; Cochran, J.B.; Tsuruta, Y.; Yusuf, N.; McKay, K.M.; Athar, M.; Timares, L.; Elmets, C.A. A murine model for the development of melanocytic nevi and their progression to melanoma. Mol. Carcinog. 2016, 55, 646–658. [Google Scholar] [CrossRef][Green Version]
- Haen, S.P.; Löffler, M.W.; Rammensee, H.G.; Brossart, P. Towards new horizons: Characterization, classification and implications of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 2020, 17, 595–610. [Google Scholar] [CrossRef]
- Castle, J.C.; Uduman, M.; Pabla, S.; Stein, R.B.; Buell, J.S. Mutation-Derived Neoantigens for Cancer Immunotherapy. Front Immunol. 2019, 10, 1856. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Hollingsworth, R.E.; Jansen, K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 2019, 4, 7. [Google Scholar] [CrossRef] [PubMed]
- Jou, J.; Harrington, K.J.; Zocca, M.B.; Ehrnrooth, E.; Cohen, E.E.W. The Changing Landscape of Therapeutic Cancer Vaccines-Novel Platforms and Neoantigen Identification. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 689–703. [Google Scholar] [CrossRef] [PubMed]
- Le, D.T.; Pardoll, D.M.; Jaffee, E.M. Cellular vaccine approaches. Cancer J. 2010, 16, 304–310. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Goldman, B.; DeFrancesco, L. The cancer vaccine roller coaster. Nat. Biotechnol. 2009, 27, 129–139. [Google Scholar] [CrossRef]
- Srivatsan, S.; Patel, J.M.; Bozeman, E.N.; Imasuen, I.E.; He, S.; Daniels, D.; Selvaraj, P. Allogeneic tumor cell vaccines the promise and limitations in clinical trials. Hum. Vacc. Immunother. 2014, 10, 52–63. [Google Scholar] [CrossRef][Green Version]
- Nasti, T.H.; Rudemiller, K.J.; Cochran, J.B.; Kim, H.K.; Tsuruta, Y.; Fineberg, N.S.; Athar, M.; Elmets, C.A.; Timares, L. Immunoprevention of chemical carcinogenesis through early recognition of oncogene mutations. J. Immunol. 2015, 194, 2683–2695. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Saxena, M.; van der Burg, S.H.; Melief, C.J.M.; Bhardwaj, N. Therapeutic cancer vaccines. Nat. Rev. Cancer 2021, 21, 360–378. [Google Scholar] [CrossRef]
- Guallar-Garrido, S.; Julián, E. Bacillus Calmette-Guérin (BCG) Therapy for Bladder Cancer: An Update. ImmunoTargets Ther. 2020, 9, 1–11. [Google Scholar] [CrossRef][Green Version]
- Redelman-Sidi, G.; Glickman, M.S.; Bochner, B.H. The mechanism of action of BCG therapy for bladder cancer—A current perspective. Nat. Rev. Urol. 2014, 11, 153–162. [Google Scholar] [CrossRef] [PubMed]
- Platten, M.; Bunse, L.; Wick, A.; Bunse, T.; Le Cornet, L.; Harting, I.; Sahm, F.; Sanghvi, K.; Tan, C.L.; Poschke, I.; et al. A vaccine targeting mutant IDH1 in newly diagnosed glioma. Nature 2021, 592, 463–468. [Google Scholar] [CrossRef] [PubMed]
- Liau, L.M.; Ashkan, K.; Tran, D.D.; Campian, J.L.; Trusheim, J.E.; Cobbs, C.S.; Heth, J.A.; Salacz, M.; Taylor, S.; D’Andre, S.D.; et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J. Transl. Med. 2018, 16, 142. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Pallerla, S.; Abdul, A.; Comeau, J.; Jois, S. Cancer Vaccines, Treatment of the Future: With Emphasis on HER2-Positive Breast Cancer. Int. J. Mol. Sci. 2021, 22, 779. [Google Scholar] [CrossRef]
- Sharma, A.; Koldovsky, U.; Xu, S.; Mick, R.; Roses, R.; Fitzpatrick, E.; Weinstein, S.; Nisenbaum, H.; Levine, B.L.; Fox, K.; et al. HER-2 pulsed dendritic cell vaccine can eliminate HER-2 expression and impact ductal carcinoma in situ. Cancer 2012, 118, 4354–4362. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Rini, B.I.; Weinberg, V.; Fong, L.; Conry, S.; Hershberg, R.M.; Small, E.J. Combination immunotherapy with prostatic acid phosphatase pulsed antigen-presenting cells (Provenge) plus bevacizumab in patients with serologic progression of prostate cancer after definitive local therapy. Cancer 2006, 107, 67–74. [Google Scholar] [CrossRef]
- Wargowski, E.; Johnson, L.E.; Eickhoff, J.C.; Delmastro, L.; Staab, M.J.; Liu, G.; McNeel, D.G. Prime-boost vaccination targeting prostatic acid phosphatase (PAP) in patients with metastatic castration-resistant prostate cancer (mCRPC) using Sipuleucel-T and a DNA vaccine. J. Immunother. Cancer 2018, 6, 21. [Google Scholar] [CrossRef][Green Version]
- Liu, K.J.; Chao, T.Y.; Chang, J.Y.; Cheng, A.L.; Ch’ang, H.J.; Kao, W.Y.; Wu, Y.C.; Yu, W.L.; Chung, T.R.; Whang-Peng, J. A phase I clinical study of immunotherapy for advanced colorectal cancers using carcinoembryonic antigen-pulsed dendritic cells mixed with tetanus toxoid and subsequent IL-2 treatment. J. Biomed. Sci. 2016, 23, 64. [Google Scholar] [CrossRef][Green Version]
- Gao, T.; Cen, Q.; Lei, H. A review on development of MUC1-based cancer vaccine. Biomed. Pharm. 2020, 132, 110888. [Google Scholar] [CrossRef]
- Corulli, L.R.; Cecil, D.L.; Gad, E.; Koehnlein, M.; Coveler, A.L.; Childs, J.S.; Lubet, R.A.; Disis, M.L. Multi-Epitope-Based Vaccines for Colon Cancer Treatment and Prevention. Front Immunol. 2021, 12, 729809. [Google Scholar] [CrossRef]
- Faiena, I.; Comin-Anduix, B.; Berent-Maoz, B.; Bot, A.; Zomorodian, N.; Sachdeva, A.; Said, J.; Cheung-Lau, G.; Pang, J.; Macabali, M.; et al. A Phase I, Open-label, Dose-escalation, and Cohort Expansion Study to Evaluate the Safety and Immune Response to Autologous Dendritic Cells Transduced with AdGMCA9 (DC-AdGMCAIX) in Patients with Metastatic Renal Cell Carcinoma. J. Immunother. 2020, 43, 273–282. [Google Scholar] [CrossRef]
- Obara, W.; Karashima, T.; Takeda, K.; Kato, R.P.; Kato, Y.; Kanehira, M.; Takata, R.; Inoue, K.; Katagiri, T.; Shuin, T.; et al. Effective induction of cytotoxic T cells recognizing an epitope peptide derived from hypoxia-inducible protein 2 (HIG2) in patients with metastatic renal cell carcinoma. Cancer Immunol. Immun. 2017, 66, 17–24. [Google Scholar] [CrossRef][Green Version]
- Yoshimura, K.; Uemura, H. Role of vaccine therapy for renal cell carcinoma in the era of targeted therapy. Int. J. Urol. 2013, 20, 744–755. [Google Scholar] [CrossRef][Green Version]
- Leroux-Roels, G. Old and new adjuvants for hepatitis B vaccines. Med. Microbiol. Immun. 2015, 204, 69–78. [Google Scholar] [CrossRef]
- Lorenzo-Luaces, P.; Sanchez, L.; Saavedra, D.; Crombet, T.; Van der Elst, W.; Alonso, A.; Molenberghs, G.; Lage, A. Identifying predictive biomarkers of CIMAvaxEGF success in non-small cell lung cancer patients. BMC Cancer 2020, 20. [Google Scholar] [CrossRef]
- Hu, Z.T.; Leet, D.E.; Allesoe, R.L.; Oliveira, G.; Li, S.Q.; Luoma, A.M.; Liu, J.Y.; Forman, J.; Huang, T.; Iorgulescu, J.B.; et al. Personal neoantigen vaccines induce persistent memory T cell responses and epitope spreading in patients with melanoma. Nat. Med. 2021, 27, 515–525. [Google Scholar] [CrossRef]
- Lei, J.; Ploner, A.; Elfstrom, K.M.; Wang, J.; Roth, A.; Fang, F.; Sundstrom, K.; Dillner, J.; Sparen, P. HPV Vaccination and the Risk of Invasive Cervical Cancer. N. Engl. J. Med. 2020, 383, 1340–1348. [Google Scholar] [CrossRef]
- Sankaranarayanan, R.; Joshi, S.; Muwonge, R.; Esmy, P.O.; Basu, P.; Prabhu, P.; Bhatla, N.; Nene, B.M.; Shaw, J.; Poli, U.R.R.; et al. Can a single dose of human papillomavirus (HPV) vaccine prevent cervical cancer? Early findings from an Indian study. Vaccine 2018, 36, 4783–4791. [Google Scholar] [CrossRef]
- Wang, M.; Zhao, J.; Zhang, L.; Wei, F.; Lian, Y.; Wu, Y.; Gong, Z.; Zhang, S.; Zhou, J.; Cao, K.; et al. Role of tumor microenvironment in tumorigenesis. J. Cancer 2017, 8, 761–773. [Google Scholar] [CrossRef]
- Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
- Nasti, T.H.; Yusuf, N.; Sherwani, M.A.; Athar, M.; Timares, L.; Elmets, C.A. Regulatory T Cells Play an Important Role in the Prevention of Murine Melanocytic Nevi and Melanomas. Cancer Prev. Res. 2021, 14, 165–174. [Google Scholar] [CrossRef]
- Kim, C.G.; Kim, C.; Yoon, S.E.; Kim, K.H.; Choi, S.J.; Kang, B.; Kim, H.R.; Park, S.H.; Shin, E.C.; Kim, Y.Y.; et al. Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. J. Hepatol. 2021, 74, 350–359. [Google Scholar] [CrossRef] [PubMed]
- Welters, M.J.; van der Sluis, T.C.; van Meir, H.; Loof, N.M.; van Ham, V.J.; van Duikeren, S.; Santegoets, S.J.; Arens, R.; de Kam, M.L.; Cohen, A.F.; et al. Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci. Transl. Med. 2016, 8, 334ra352. [Google Scholar] [CrossRef]
- Thommen, D.S.; Schumacher, T.N. T Cell Dysfunction in Cancer. Cancer Cell 2018, 33, 547–562. [Google Scholar] [CrossRef][Green Version]
- Kim, M.H.; Kim, C.G.; Kim, S.K.; Shin, S.J.; Choe, E.A.; Park, S.H.; Shin, E.C.; Kim, J. YAP-Induced PD-L1 Expression Drives Immune Evasion in BRAFi-Resistant Melanoma. Cancer Immunol. Res. 2018, 6, 255–266. [Google Scholar] [CrossRef][Green Version]
- Chao, M.P.; Majeti, R.; Weissman, I.L. Programmed cell removal: A new obstacle in the road to developing cancer. Nat. Rev. Cancer 2011, 12, 58–67. [Google Scholar] [CrossRef] [PubMed]
- Ford, K.; Hanley, C.J.; Mellone, M.; Szyndralewiez, C.; Heitz, F.; Wiesel, P.; Wood, O.; Machado, M.; Lopez, M.A.; Ganesan, A.P.; et al. NOX4 Inhibition Potentiates Immunotherapy by Overcoming Cancer-Associated Fibroblast-Mediated CD8 T-cell Exclusion from Tumors. Cancer Res. 2020, 80, 1846–1860. [Google Scholar] [CrossRef][Green Version]
- Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef] [PubMed]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Canellas, A.; Hernando-Momblona, X.; et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Kim, C.G.; Jang, M.; Kim, Y.; Leem, G.; Kim, K.H.; Lee, H.; Kim, T.S.; Choi, S.J.; Kim, H.D.; Han, J.W.; et al. VEGF-A drives TOX-dependent T cell exhaustion in anti-PD-1-resistant microsatellite stable colorectal cancers. Sci. Immunol. 2019, 4. [Google Scholar] [CrossRef]
- Saxena, M.; Bhardwaj, N. Re-Emergence of Dendritic Cell Vaccines for Cancer Treatment. Trends Cancer 2018, 4, 119–137. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef][Green Version]
- Seidel, J.A.; Otsuka, A.; Kabashima, K. Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Front. Oncol. 2018, 8, 86. [Google Scholar] [CrossRef]
- Maleki Vareki, S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J. Immunother. Cancer 2018, 6, 157. [Google Scholar] [CrossRef]
- Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 2010, 33, 828–833. [Google Scholar] [CrossRef]
- Beer, T.M.; Kwon, E.D.; Drake, C.G.; Fizazi, K.; Logothetis, C.; Gravis, G.; Ganju, V.; Polikoff, J.; Saad, F.; Humanski, P.; et al. Randomized, Double-Blind, Phase III Trial of Ipilimumab Versus Placebo in Asymptomatic or Minimally Symptomatic Patients With Metastatic Chemotherapy-Naive Castration-Resistant Prostate Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 40–47. [Google Scholar] [CrossRef]
- Curran, M.A.; Glisson, B.S. New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation. Annu. Rev. Med. 2019, 70, 409–424. [Google Scholar] [CrossRef]
- Fong, L.; Carroll, P.; Weinberg, V.; Chan, S.; Lewis, J.; Corman, J.; Amling, C.L.; Stephenson, R.A.; Simko, J.; Sheikh, N.A.; et al. Activated lymphocyte recruitment into the tumor microenvironment following preoperative sipuleucel-T for localized prostate cancer. J. Natl. Cancer Inst. 2014, 106, dju268. [Google Scholar] [CrossRef]
- Gulley, J.L.; Madan, R.A.; Tsang, K.Y.; Jochems, C.; Marté, J.L.; Farsaci, B.; Tucker, J.A.; Hodge, J.W.; Liewehr, D.J.; Steinberg, S.M.; et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol. Res. 2014, 2, 133–141. [Google Scholar] [CrossRef][Green Version]
- Brossart, P. The Role of Antigen Spreading in the Efficacy of Immunotherapies. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 4442–4447. [Google Scholar] [CrossRef]
- Kim, C.G.; Sang, Y.B.; Lee, J.H.; Chon, H.J. Combining Cancer Vaccines with Immunotherapy: Establishing a New Immunological Approach. Int. J. Mol. Sci. 2021, 22, 8035. [Google Scholar] [CrossRef] [PubMed]
- Ahmadzadeh, M.; Johnson, L.; Heemskerk, B.; Wunderlich, J.; Rosenberg, S. Tumor Antigen-specific CD8 T Cells Infiltrating the Tumor Express High Levels of PD-1 and are Functionally Impaired. Clin. Immunol. 2009, 131, S38. [Google Scholar] [CrossRef]
- Thompson, R.H.; Dong, H.D.; Lohse, C.M.; Leibovich, B.C.; Blute, M.L.; Cheville, J.C.; Kwon, E.D. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin. Cancer Res. 2007, 13, 1757–1761. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Baxter, A.G.; Hodgkin, P.D. Activation rules: The two-signal theories of immune activation. Nat. Rev. Immunol. 2002, 2, 439–446. [Google Scholar] [CrossRef]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef][Green Version]
- Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Urger, M.C.B.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8(+) T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef]
- Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef][Green Version]
- Wang, W.S.; Lau, R.; Yu, D.H.; Zhu, W.W.; Korman, A.; Weber, J. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4(+)CD25(Hi) regulatory T cells. Int. Immunol. 2009, 21, 1065–1077. [Google Scholar] [CrossRef][Green Version]
- Khoja, L.; Day, D.; Wei-Wu Chen, T.; Siu, L.L.; Hansen, A.R. Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: A systematic review. Ann. Oncol. 2017, 28, 2377–2385. [Google Scholar] [CrossRef]
- De Velasco, G.; Je, Y.; Bossé, D.; Awad, M.M.; Ott, P.A.; Moreira, R.B.; Schutz, F.; Bellmunt, J.; Sonpavde, G.P.; Hodi, F.S.; et al. Comprehensive Meta-analysis of Key Immune-Related Adverse Events from CTLA-4 and PD-1/PD-L1 Inhibitors in Cancer Patients. Cancer Immunol. Res. 2017, 5, 312–318. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Soares, K.C.; Rucki, A.A.; Wu, A.A.; Olino, K.; Xiao, Q.; Chai, Y.; Wamwea, A.; Bigelow, E.; Lutz, E.; Liu, L.; et al. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J. Immunother. 2015, 38, 1–11. [Google Scholar] [CrossRef][Green Version]
- Antonios, J.P.; Soto, H.; Everson, R.G.; Orpilla, J.; Moughon, D.; Shin, N.; Sedighim, S.; Yong, W.H.; Li, G.; Cloughesy, T.F.; et al. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight 2016, 1, e87059. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Rice, A.E.; Latchman, Y.E.; Balint, J.P.; Lee, J.H.; Gabitzsch, E.S.; Jones, F.R. An HPV-E6/E7 immunotherapy plus PD-1 checkpoint inhibition results in tumor regression and reduction in PD-L1 expression. Cancer Gene 2015, 22, 454–462. [Google Scholar] [CrossRef][Green Version]
- Quezada, S.A.; Peggs, K.S.; Curran, M.A.; Allison, J.P. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J. Clin. Investig. 2006, 116, 1935–1945. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Wada, S.; Jackson, C.M.; Yoshimura, K.; Yen, H.R.; Getnet, D.; Harris, T.J.; Goldberg, M.V.; Bruno, T.C.; Grosso, J.F.; Durham, N.; et al. Sequencing CTLA-4 blockade with cell-based immunotherapy for prostate cancer. J. Transl. Med. 2013, 11, 89. [Google Scholar] [CrossRef][Green Version]
- 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, 116. [Google Scholar] [CrossRef]
- Zhao, J.; Chen, Y.; Ding, Z.Y.; Liu, J.Y. Safety and Efficacy of Therapeutic Cancer Vaccines Alone or in Combination with Immune Checkpoint Inhibitors in Cancer Treatment. Front. Pharmacol. 2019, 10, 1184. [Google Scholar] [CrossRef]
- Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell 2017, 170, 1109–1119.e10. [Google Scholar] [CrossRef][Green Version]
- Harrington, K.J.; Kong, A.; Mach, N.; Chesney, J.A.; Fernandez, B.C.; Rischin, D.; Cohen, E.E.W.; Radcliffe, H.S.; Gumuscu, B.; Cheng, J.; et al. Talimogene Laherparepvec and Pembrolizumab in Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (MASTERKEY-232): A Multicenter, Phase 1b Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 5153–5161. [Google Scholar] [CrossRef]
- Weber, J.S.; Kudchadkar, R.R.; Yu, B.; Gallenstein, D.; Horak, C.E.; Inzunza, H.D.; Zhao, X.; Martinez, A.J.; Wang, W.; Gibney, G.; et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2013, 31, 4311–4318. [Google Scholar] [CrossRef][Green Version]
- Puzanov, I.; Milhem, M.M.; Minor, D.; Hamid, O.; Li, A.; Chen, L.; Chastain, M.; Gorski, K.S.; Anderson, A.; Chou, J.; et al. Talimogene Laherparepvec in Combination With Ipilimumab in Previously Untreated, Unresectable Stage IIIB-IV Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 2619–2626. [Google Scholar] [CrossRef][Green Version]
- Ku, J.; Wilenius, K.; Larsen, C.; Guzman, K.D.; Yoshinaga, S.; Turner, J.S.; Lam, R.Y.; Scholz, M.C. Survival after sipuleucel-T (SIP-T) and low-dose ipilimumab (IPI) in men with metastatic, progressive, castrate-resistant prostate cancer (M-CRPC). J. Clin. Oncol. 2018, 36, 368. [Google Scholar] [CrossRef]
- Van den Eertwegh, A.J.; Versluis, J.; van den Berg, H.P.; Santegoets, S.J.; van Moorselaar, R.J.; van der Sluis, T.M.; Gall, H.E.; Harding, T.C.; Jooss, K.; Lowy, I.; et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: A phase 1 dose-escalation trial. Lancet. Oncol. 2012, 13, 509–517. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Ott, P.A.; Hu-Lieskovan, S.; Chmielowski, B.; Govindan, R.; Naing, A.; Bhardwaj, N.; Margolin, K.; Awad, M.M.; Hellmann, M.D.; Lin, J.J.; et al. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients with Advanced Melanoma, Non-small Cell Lung Cancer, or Bladder Cancer. Cell 2020, 183, 347–362.e24. [Google Scholar] [CrossRef]
- Hau, M.; Schwartz, K.L.; Frenette, C.; Mogck, I.; Gubbay, J.B.; Severini, A.; Hiebert, J.; Deeks, S.L.; Morris, S.K. Local public health response to vaccine-associated measles: Case report. BMC Public Health 2013, 13, 269. [Google Scholar] [CrossRef][Green Version]
- Pergam, S.A.; Englund, J.A.; Kamboj, M.; Gans, H.A.; Young, J.H.; Hill, J.A.; Savani, B.; Chemaly, R.F.; Dadwal, S.S.; Storek, J.; et al. Preventing Measles in Immunosuppressed Cancer and Hematopoietic Cell Transplantation Patients: A Position Statement by the American Society for Transplantation and Cellular Therapy. Biol. Blood Marrow Transpl. 2019, 25, e321–e330. [Google Scholar] [CrossRef]
- Tamma, P. Vaccines in immunocompromised patients. Pediatr Rev. 2010, 31, 38–40. [Google Scholar] [CrossRef][Green Version]
- Ariza-Heredia, E.J.; Chemaly, R.F. Practical review of immunizations in adult patients with cancer. Hum. Vaccin Immunother. 2015, 11, 2606–2614. [Google Scholar] [CrossRef][Green Version]
- Mitchell, D.A.; Batich, K.A.; Gunn, M.D.; Huang, M.N.; Sanchez-Perez, L.; Nair, S.K.; Congdon, K.L.; Reap, E.A.; Archer, G.E.; Desjardins, A.; et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature 2015, 519, 366–369. [Google Scholar] [CrossRef]
- Shekarian, T.; Sivado, E.; Jallas, A.C.; Depil, S.; Kielbassa, J.; Janoueix-Lerosey, I.; Hutter, G.; Goutagny, N.; Bergeron, C.; Viari, A.; et al. Repurposing rotavirus vaccines for intratumoral immunotherapy can overcome resistance to immune checkpoint blockade. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Läubli, H.; Balmelli, C.; Kaufmann, L.; Stanczak, M.; Syedbasha, M.; Vogt, D.; Hertig, A.; Müller, B.; Gautschi, O.; Stenner, F.; et al. Influenza vaccination of cancer patients during PD-1 blockade induces serological protection but may raise the risk for immune-related adverse events. J. Immunother. Cancer 2018, 6, 40. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Spagnolo, F.; Boutros, A.; Croce, E.; Cecchi, F.; Arecco, L.; Tanda, E.; Pronzato, P.; Lambertini, M. Influenza vaccination in cancer patients receiving immune checkpoint inhibitors: A systematic review. Eur. J. Clin. Investig. 2021, 51, e13604. [Google Scholar] [CrossRef] [PubMed]
- Gopalakrishnan, R.; Johnson, D.B.; York, S.; Neuss, M.N.; Osterman, T.J.; Chism, D.D.; Ancell, K.K.; Mayer, I.A.; Abramson, V.G.; Levy, M.A.; et al. Impact of the influenza vaccination on cancer patients undergoing therapy with immune checkpoint inhibitors (ICI). J. Clin. Oncol. 2018, 36, 3053. [Google Scholar] [CrossRef]
- Awadalla, M.; Golden, D.L.A.; Mahmood, S.S.; Alvi, R.M.; Mercaldo, N.D.; Hassan, M.Z.O.; Banerji, D.; Rokicki, A.; Mulligan, C.; Murphy, S.P.T.; et al. Influenza vaccination and myocarditis among patients receiving immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 53. [Google Scholar] [CrossRef]
- Bersanelli, M.; Giannarelli, D.; De Giorgi, U.; Pignata, S.; Di Maio, M.; Clemente, A.; Verzoni, E.; Giusti, R.; Di Napoli, M.; Aprile, G.; et al. INfluenza Vaccine Indication During therapy with Immune checkpoint inhibitors: A multicenter prospective observational study (INVIDIa-2). J. Immunother Cancer 2021, 9, e002619. [Google Scholar] [CrossRef]
- Waissengrin, B.; Agbarya, A.; Safadi, E.; Padova, H.; Wolf, I. Short-term safety of the BNT162b2 mRNA COVID-19 vaccine in patients with cancer treated with immune checkpoint inhibitors. Lancet. Oncol. 2021, 22, 581–583. [Google Scholar] [CrossRef]
- Terpos, E.; Zagouri, F.; Liontos, M.; Sklirou, A.D.; Koutsoukos, K.; Markellos, C.; Briasoulis, A.; Papanagnou, E.D.; Trougakos, I.P.; Dimopoulos, M.A. Low titers of SARS-CoV-2 neutralizing antibodies after first vaccination dose in cancer patients receiving checkpoint inhibitors. J. Hematol. Oncol. 2021, 14, 86. [Google Scholar] [CrossRef]
- Lasagna, A.; Agustoni, F.; Percivalle, E.; Borgetto, S.; Paulet, A.; Comolli, G.; Sarasini, A.; Bergami, F.; Sammartino, J.C.; Ferrari, A.; et al. A snapshot of the immunogenicity, efficacy and safety of a full course of BNT162b2 anti-SARS-CoV-2 vaccine in cancer patients treated with PD-1/PD-L1 inhibitors: A longitudinal cohort study. ESMO Open 2021, 6, 100272. [Google Scholar] [CrossRef]
- Thakkar, A.; Gonzalez-Lugo, J.D.; Goradia, N.; Gali, R.; Shapiro, L.C.; Pradhan, K.; Rahman, S.; Kim, S.Y.; Ko, B.; Sica, R.A.; et al. Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell 2021, 39, 1081–1090.e2. [Google Scholar] [CrossRef] [PubMed]
Cancer Type | Vaccine | Description | Type | Mechanism | Stage of Development | References |
---|---|---|---|---|---|---|
Bladder | BCG (Bacillus Calmette–Guérin) | Autologous (was used mainly against tuberculosis) | Therapeutic | Internalization of BCG antigen and activation of antigen-specific CD8 and CD4 T cells; direct cytotoxicity of the tumor cells | In clinical use | Reviewed in [21,22] |
Brain | IDH1(R132H)-specific peptide vaccine | Allogenic (Isocitrate dehydrogenase1, gets mutated in gliomas) | Therapeutic | Specific immune response against the mutated protein | Phase 1 | [23] |
DCVax®-L (Dendrtitic cell based personalized vaccine) | Autologous | Therapeutic | Patient-derived dendritic cells are labeled with patient’s tumor cells and injected intradermal to induce an immune response | Phase 3 | [24] | |
Breast | Her 2 directed Cellular/DNA/viral | Autologous or allogenic | Therapeutic | Activation of immune response | Phase 1/2 | Many Phase1/phase 2 trials reviewed in [25] |
h tert (telomerase reverse transcriptase) | Autologous or allogenic | Therapeutic | Activation of CTLs against mutations in overexpressing breast cancer cells | Reviewed in [26]. | ||
Prostate | Sipuleucel-T (prostate acid phosphatase antigen (PAP)) | Autologous (patients APCs incubated with PAP and GM-CSF) | Therapeutic | T cell | In clinical use | [27,28] |
Colorectal | CEA (carcinoembryonic antigen) Muc1 Peptide/DNA | Autologous Allogenic Allogenic | Therapeutic | CTL response | Preclinical, In clinical trial | [29,30,31] |
Kidney | carbonic-anhydrase IX HLA-A 0201/0206-restricted epitope peptide (HIG2-9-4) vaccine | Allogenic/ Autologous Autologous | Therapeutic Therapeutic | Increase in IFN responses CTL CTL | In clinical trials | [32,33,34] |
Liver | HEPLISAV-B Heptatis B surface antigen | Allogenic | Preventive | antibody response CTL | In clinical use | [35] |
Lung | CIMAvax-EGF | Allogenic | Preventive | antibody | Clinical trial | [36] |
Melanoma | Neovax (personalized neoantigens) | Autologous | Preventive (after surgery) | CD4 and CD8 | Clinical trial | [37] |
Cervical | Gardasil4/9 Cervarix–contain L1 proteins from different strains | Allogenic | Preventive | Mainly induces neutralizing antibodies against various strains of HPV | In clinical use | [38,39] |
Cancer Type | Vaccine | ICI Agent | Type | Summary | Reference |
---|---|---|---|---|---|
Melanoma | Talimogene Laherparepvec (GM-CSF) | Pembrolizumab | Phase III | Well tolerated (only grade 1 and 2 toxicities) and showed OR = 62% | [80] |
Head and Neck | Talimogene Laherparepvec (GM-CSF) | Pembrolizumab | Phase 1 | irAEs > 50%, related to either GM-CSF or Pembrolizumab Only 13% partial response | [81] |
Melanoma | gp100280-288 (288 V), and NY-ESO-1157-165 (165 V). peptide vaccine | Nivolumab | Phase 1 | Well tolerated: 53% had disease progression at 2 years. Progression was associated with increased regulatory T cells and a decrease in antigen-specific CD8 T cells | [82] |
Melanoma | Talimogene Laherparepvec (GM-CSF) | Ipilimumab | Phase 1 | Well tolerated, Grade 3 and 4 irAEs: 26%. Objective response: 50% | [83] |
Prostate cancer | Sipuleucel-T (SIP-T) | Ipilimumab | Phase III | Adverse effects negligible. Median survival < 4 years | [84] |
Prostate cancer | GVAX | ipilimumab | Phase I | Well tolerated. 50% reduction in prostate-specific antigen in the combination group | [85] |
Metastatic melanoma | Gp100 peptide vaccine | Ipilimumab | Phase III | Well tolerated. Grade 3 and 4 irAEs: 10–15%. Overall survival: 10 months for combination vs. 6 months for GP100 alone. No difference between ipi and combination | [86] |
Melanoma/bladder and lung cancer | Neo-PV-01 (personalized neoantigens) | Nivolumab | Phase IIb | Safe, efficacious and activation of CD4 and CD8 T Cells | [87] |
Cancer Type | Vaccine | Patient Number | Safety | Efficacy | Reference |
---|---|---|---|---|---|
Lung | Influenza | Vaccinated; n = 23 Healthy: n = 11 control cancer: n = 40 | irAEs 52.2% vs. 25.5% | Similar humoral response in healthy or cancer patients | [94] |
Different cancer types | Influenza | n = 1124 | Any grade irAEs 28.9%, grade 3–4: 7.5% | High antibody titers, CTLs, very few patients experienced Influenza infection | [95] |
Different cancer types | Influenza | n = 1188 vaccinated n = 581 unvaccinated n = 607 | Vaccine related adverse events: 1.5% grades 1–2 | Similar incidence of influenza-like illness, Fatality 4.3% (unvaccinated) vs. 0% (vaccinated) | [98] |
Many types, 50% lung cancer | COVID-19 | n = 134 | No Vaccine related toxicities | NA | [99] |
Different cancer types; Lung and bladder 25% each | COVID-19 | n = 59 (cancer patients) n = 283 (controls) | Only one patient had irAEs | Neutralizing antibody titers 22% vs. 38% in controls | [100] |
Different types; Lung cancer 76% | COVID-19 | n = 88 | Fever and pain at injection site. 1 patient with grade 3 irAEs. | High seropositivity, and CD8 and CD4 responses | [101] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Nasti, T.H.; Eberhardt, C.S. Vaccination against Cancer or Infectious Agents during Checkpoint Inhibitor Therapy. Vaccines 2021, 9, 1396. https://doi.org/10.3390/vaccines9121396
Nasti TH, Eberhardt CS. Vaccination against Cancer or Infectious Agents during Checkpoint Inhibitor Therapy. Vaccines. 2021; 9(12):1396. https://doi.org/10.3390/vaccines9121396
Chicago/Turabian StyleNasti, Tahseen H., and Christiane S. Eberhardt. 2021. "Vaccination against Cancer or Infectious Agents during Checkpoint Inhibitor Therapy" Vaccines 9, no. 12: 1396. https://doi.org/10.3390/vaccines9121396