Immune Specific and Tumor-Dependent mRNA Vaccines for Cancer Immunotherapy: Reprogramming Clinical Translation into Tumor Editing Therapy
Abstract
:1. Introduction
2. mRNA Vaccine Pharmacology
3. Optimization of mRNA Translation
4. Immunogenicity of mRNA Vaccines
5. mRNA Vaccine Efficacy
6. Tumor Intrinsic Resistance
7. Tumor Extrinsic Resistance
8. Cell-Based Cancer Vaccines
9. Peptide-Based Cancer Vaccines
10. Nucleic Acid-Based Cancer Vaccines
11. Viral-Based Cancer Vaccines
12. Lipid mRNA-Based Nanovaccines
13. Dendritic Cell mRNA-Based Vaccines
14. Challenges and Future Perspectives
15. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
References
- Szabó, G.T.; Mahiny, A.J.; Vlatkovic, I. COVID-19 mRNA vaccines: Platforms and current developments. Mol. Ther. 2022, 30, 1850–1868. [Google Scholar] [CrossRef]
- Chakraborty, C.; Sharma, A.R.; Bhattacharya, M.; Lee, S.S. From COVID-19 to Cancer mRNA Vaccines: Moving from Bench to Clinic in the Vaccine Landscape. Front. Immunol. 2021, 12, 679344. [Google Scholar] [CrossRef]
- Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines—A new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef]
- Verbeke, R.; Hogan, M.J.; Loré, K.; Pardi, N. Innate immune mechanisms of mRNA vaccines. Immunity 2022, 55, 1993–2005. [Google Scholar] [CrossRef]
- Lorentzen, C.L.; Haanen, J.B.; Met, Ö.; Svane, I.M. Clinical advances and ongoing trials on mRNA vaccines for cancer treatment. Lancet Oncol. 2022, 23, e450–e458. [Google Scholar] [CrossRef]
- Adamik, J.; Butterfield, L.H. What’s next for cancer vaccines. Sci. Transl. Med. 2022, 14, eabo4632. [Google Scholar] [CrossRef]
- Lopes, A.; Vandermeulen, G.; Préat, V. Cancer DNA vaccines: Current preclinical and clinical developments and future perspectives. J. Exp. Clin. Cancer Res. 2019, 38, 146. [Google Scholar] [CrossRef]
- Liang, W.; Lin, Z.; Du, C.; Qiu, D.; Zhang, Q. mRNA modification orchestrates cancer stem cell fate decisions. Mol. Cancer 2020, 19, 38. [Google Scholar] [CrossRef]
- Jia, L.; Mao, Y.; Ji, Q.; Dersh, D.; Yewdell, J.W.; Qian, S.B. Decoding mRNA translatability and stability from the 5’ UTR. Nat. Struct. Mol. Biol. 2020, 27, 814–821. [Google Scholar] [CrossRef] [PubMed]
- Presnyak, V.; Alhusaini, N.; Chen, Y.H.; Martin, S.; Morris, N.; Kline, N.; Olson, S.; Weinberg, D.; Baker, K.E.; Graveley, B.R.; et al. Codon optimality is a major determinant of mRNA stability. Cell 2015, 160, 1111–1124. [Google Scholar] [CrossRef] [PubMed]
- Bidram, M.; Zhao, Y.; Shebardina, N.G.; Baldin, A.V.; Bazhin, A.V.; Ganjalikhany, M.R.; Zamyatnin, A.A., Jr. Ganjalikhani-Hakemi, M. mRNA-Based Cancer Vaccines: A Therapeutic Strategy for the Treatment of Melanoma Patients. Vaccines 2021, 9, 1060. [Google Scholar] [CrossRef]
- Liu, C.; Shi, Q.; Huang, X.; Koo, S.; Kong, N.; Tao, W. mRNA-based cancer therapeutics. Nat. Rev. Cancer 2023, 23, 526–543. [Google Scholar] [CrossRef]
- Verbeke, R.; Lentacker, I.; De Smedt, S.C.; Dewitte, H. Three decades of messenger RNA vaccine development. Nano Today 2019, 28, 100766. [Google Scholar] [CrossRef]
- Huang, X.; Kong, N.; Zhang, X.; Cao, Y.; Langer, R.; Tao, W. The landscape of mRNA nanomedicine. Nat. Med. 2022, 28, 2273–2287. [Google Scholar] [CrossRef]
- Heine, A.; Juranek, S.; Brossart, P. Clinical and immunological effects of mRNA vaccines in malignant diseases. Mol. Cancer 2021, 20, 52. [Google Scholar] [CrossRef]
- Fu, J.; Chen, F.; Lin, Y.; Gao, J.; Chen, A.; Yang, J. Discovery and characterization of tumor antigens in hepatocellular carcinoma for mRNA vaccine development. J. Cancer Res. Clin. Oncol. 2023, 149, 4047–4061. [Google Scholar] [CrossRef]
- Hassett, K.J.; Benenato, K.E.; Jacquinet, E.; Lee, A.; Woods, A.; Yuzhakov, O.; Himansu, S.; Deterling, J.; Geilich, B.M.; Ketova, T.; et al. Optimization of Lipid Nanoparticles for Intramuscular Administration of mRNA Vaccines. Mol. Ther. Nucleic Acids 2019, 15, 1–11. [Google Scholar] [CrossRef]
- Rohner, E.; Yang, R.; Foo, K.S.; Goedel, A.; Chien, K.R. Unlocking the promise of mRNA therapeutics. Nat. Biotechnol. 2022, 40, 1586–1600. [Google Scholar] [CrossRef]
- Bae, H.; Coller, J. Codon optimality-mediated mRNA degradation: Linking translational elongation to mRNA stability. Mol. Cell 2022, 82, 1467–1476. [Google Scholar] [CrossRef]
- Schlake, T.; Thran, M.; Fiedler, K.; Heidenreich, R.; Petsch, B.; Fotin-Mleczek, M. mRNA: A Novel Avenue to Antibody Therapy? Mol. Ther. 2019, 27, 773–784. [Google Scholar] [CrossRef]
- Liu, J.; Fu, M.; Wang, M.; Wan, D.; Wei, Y.; Wei, X. Cancer vaccines as promising immuno-therapeutics: Platforms and current progress. J. Hematol. Oncol. 2022, 15, 28. [Google Scholar] [CrossRef]
- Hanson, G.; Coller, J. Codon optimality, bias and usage in translation and mRNA decay. Nat. Rev. Mol. Cell. Biol. 2018, 19, 20–30. [Google Scholar] [CrossRef]
- Eralp, Y. Application of mRNA Technology in Cancer Therapeutics. Vaccines 2022, 10, 1262. [Google Scholar] [CrossRef]
- Teijaro, J.R.; Farber, D.L. COVID-19 vaccines: Modes of immune activation and future challenges. Nat. Rev. Immunol. 2021, 21, 195–197. [Google Scholar] [CrossRef] [PubMed]
- Gilboa, E.; Boczkowski, D.; Nair, S.K. The Quest for mRNA Vaccines. Nucleic Acid Ther. 2022, 32, 449–456. [Google Scholar] [CrossRef]
- Ye, Z.; Harmon, J.; Ni, W.; Li, Y.; Wich, D.; Xu, Q. The mRNA Vaccine Revolution: COVID-19 Has Launched the Future of Vaccinology. ACS Nano 2023, 17, 15231–15253. [Google Scholar] [CrossRef]
- Blanchard, E.L.; Loomis, K.H.; Bhosle, S.M.; Vanover, D.; Baumhof, P.; Pitard, B.; Zurla, C.; Santangelo, P.J. Proximity Ligation Assays for In Situ Detection of Innate Immune Activation: Focus on In Vitro-Transcribed mRNA. Mol. Ther. Nucleic Acids 2019, 14, 52–66. [Google Scholar] [CrossRef]
- Rosini, R.; Nicchi, S.; Pizza, M.; Rappuoli, R. Vaccines against Antimicrobial Resistance. Front. Immunol. 2020, 11, 1048. [Google Scholar] [CrossRef]
- Klugman, K.P.; Black, S. Impact of existing vaccines in reducing antibiotic resistance: Primary and secondary effects. Proc. Natl. Acad. Sci. USA 2018, 115, 12896–12901. [Google Scholar] [CrossRef]
- van Elsas, M.J.; van Hall, T.; van der Burg, S.H. Future Challenges in Cancer Resistance to Immunotherapy. Cancers 2020, 12, 935. [Google Scholar] [CrossRef]
- Zhou, J.; Ji, Q.; Li, Q. Resistance to anti-EGFR therapies in metastatic colorectal cancer: Underlying mechanisms and reversal strategies. J. Exp. Clin. Cancer Res. 2021, 40, 328. [Google Scholar] [CrossRef] [PubMed]
- Kirchhammer, N.; Trefny, M.P.; Auf der Maur, P.; Läubli, H.; Zippelius, A. Combination cancer immunotherapies: Emerging treatment strategies adapted to the tumor microenvironment. Sci. Transl. Med. 2022, 14, eabo3605. [Google Scholar] [CrossRef]
- Szakács, G.; Paterson, J.K.; Ludwig, J.A.; Booth-Genthe, C.; Gottesman, M.M. Targeting multidrug resistance in cancer. Nat. Rev. Drug Discov. 2006, 5, 219–234. [Google Scholar] [CrossRef]
- Morad, G.; Helmink, B.A.; Sharma, P.; Wargo, J.A. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell 2021, 184, 5309–5337. [Google Scholar] [CrossRef]
- Pitt, J.M.; Vétizou, M.; Daillère, R.; Roberti, M.P.; Yamazaki, T.; Routy, B.; Lepage, P.; Boneca, I.G.; Chamaillard, M.; Kroemer, G.; et al. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2016, 44, 1255–1269. [Google Scholar] [CrossRef]
- Valeri, A.; García-Ortiz, A.; Castellano, E.; Córdoba, L.; Maroto-Martín, E.; Encinas, J.; Leivas, A.; Río, P.; Martínez-López, J. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front. Immunol. 2022, 13, 953849. [Google Scholar] [CrossRef]
- Barry, S.T.; Gabrilovich, D.I.; Sansom, O.J.; Campbell, A.D.; Morton, J.P. Therapeutic targeting of tumour myeloid cells. Nat. Rev. Cancer 2023, 23, 216–237. [Google Scholar] [CrossRef]
- Saw, P.E.; Chen, J.; Song, E. Targeting CAFs to overcome anticancer therapeutic resistance. Trends Cancer 2022, 8, 527–555. [Google Scholar] [CrossRef]
- Kloosterman, D.J.; Akkari, L. Macrophages at the interface of the co-evolving cancer ecosystem. Cell 2023, 186, 1627–1651. [Google Scholar] [CrossRef]
- Christofides, A.; Strauss, L.; Yeo, A.; Cao, C.; Charest, A.; Boussiotis, V.A. The complex role of tumor-infiltrating macrophages. Nat. Immunol. 2022, 23, 1148–1156. [Google Scholar] [CrossRef]
- Cassetta, L.; Pollard, J.W. Targeting macrophages: Therapeutic approaches in cancer. Nat. Rev. Drug Discov. 2018, 17, 887–904. [Google Scholar] [CrossRef]
- O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
- Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
- Sellars, M.C.; Wu, C.J.; Fritsch, E.F. Cancer vaccines: Building a bridge over troubled waters. Cell 2022, 185, 2770–2788. [Google Scholar] [CrossRef]
- Harari, A.; Graciotti, M.; Bassani-Sternberg, M.; Kandalaft, L.E. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat. Rev. Drug Discov. 2020, 19, 635–652. [Google Scholar] [CrossRef]
- Palucka, K.; Banchereau, J. Dendritic-cell-based therapeutic cancer vaccines. Immunity 2013, 39, 38–48. [Google Scholar] [CrossRef]
- Sabado, R.L.; Balan, S.; Bhardwaj, N. Dendritic cell-based immunotherapy. Cell Res. 2017, 27, 74–95. [Google Scholar] [CrossRef]
- Gardner, A.; de Mingo Pulido, Á.; Ruffell, B. Dendritic Cells and Their Role in Immunotherapy. Front. Immunol. 2020, 11, 924. [Google Scholar] [CrossRef] [PubMed]
- Fu, C.; Zhou, L.; Mi, Q.S.; Jiang, A. Plasmacytoid Dendritic Cells and Cancer Immunotherapy. Cells 2022, 11, 222. [Google Scholar] [CrossRef] [PubMed]
- Stephens, A.J.; Burgess-Brown, N.A.; Jiang, S. Beyond Just Peptide Antigens: The Complex World of Peptide-Based Cancer Vaccines. Front. Immunol. 2021, 12, 696791. [Google Scholar] [CrossRef] [PubMed]
- Peng, M.; Mo, Y.; Wang, Y.; Wu, P.; Zhang, Y.; Xiong, F.; Guo, C.; Wu, X.; Li, Y.; Li, X.; et al. Neoantigen vaccine: An emerging tumor immunotherapy. Mol. Cancer 2019, 18, 128. [Google Scholar] [CrossRef]
- Kimura, T.; Egawa, S.; Uemura, H. Personalized peptide vaccines and their relation to other therapies in urological cancer. Nat. Rev. Urol. 2017, 14, 501–510. [Google Scholar] [CrossRef]
- Parmiani, G.; Castelli, C.; Dalerba, P.; Mortarini, R.; Rivoltini, L.; Marincola, F.M.; Anichini, A. Cancer immunotherapy with peptide-based vaccines: What have we achieved? Where are we going? J. Natl. Cancer Inst. 2002, 94, 805–818. [Google Scholar] [CrossRef] [PubMed]
- Mukherjee, A.; de Izarra, A.; Degrouard, J.; Olive, E.; Maiti, P.K.; Jang, Y.H.; Lansac, Y. Protamine-Controlled Reversible DNA Packaging: A Molecular Glue. ACS Nano 2021, 15, 13094–13104. [Google Scholar] [CrossRef]
- Ukogu, O.A.; Smith, A.D.; Devenica, L.M.; Bediako, H.; McMillan, R.B.; Ma, Y.; Balaji, A.; Schwab, R.D.; Anwar, S.; Dasgupta, M.; et al. Protamine loops DNA in multiple steps. Nucleic Acids Res. 2020, 48, 6108–6119. [Google Scholar] [CrossRef] [PubMed]
- Levy, J.H.; Ghadimi, K.; Kizhakkedathu, J.N.; Iba, T. What’s fishy about protamine? Clinical use, adverse reactions, and potential alternatives. J. Thromb. Haemost. 2023, 21, 1714–1723. [Google Scholar] [CrossRef] [PubMed]
- Bennett, R.; Yakkundi, A.; McKeen, H.D.; McClements, L.; McKeogh, T.J.; McCrudden, C.M.; Arthur, K.; Robson, T.; McCarthy, H.O. RALA-mediated delivery of FKBPL nucleic acid therapeutics. Nanomedicine 2015, 10, 2989–3001. [Google Scholar] [CrossRef]
- Chen, G.; Zhao, B.; Ruiz, E.F.; Zhang, F. Advances in the polymeric delivery of nucleic acid vaccines. Theranostics 2022, 12, 4081–4109. [Google Scholar] [CrossRef] [PubMed]
- Teplensky, M.H.; Evangelopoulos, M.; Dittmar, J.W.; Forsyth, C.M.; Sinegra, A.J.; Wang, S.; Mirkin, C.A. Multi-antigen spherical nucleic acid cancer vaccines. Nat. Biomed. Eng. 2023, 7, 911–927. [Google Scholar] [CrossRef]
- Geall, A.J.; Verma, A.; Otten, G.R.; Shaw, C.A.; Hekele, A.; Banerjee, K.; Cu, Y.; Beard, C.W.; Brito, L.A.; Krucker, T.; et al. Nonviral delivery of self-amplifying RNA vaccines. Proc. Natl. Acad. Sci. USA 2012, 109, 14604–14609. [Google Scholar] [CrossRef]
- Desmet, C.J.; Ishii, K.J. Nucleic acid sensing at the interface between innate and adaptive immunity in vaccination. Nat. Rev. Immunol. 2012, 12, 479–491. [Google Scholar] [CrossRef]
- Iurescia, S.; Fioretti, D.; Rinaldi, M. Targeting Cytosolic Nucleic Acid-Sensing Pathways for Cancer Immunotherapies. Front. Immunol. 2018, 9, 711. [Google Scholar] [CrossRef] [PubMed]
- Beaudoin, C.A.; Bartas, M.; Volná, A.; Pečinka, P.; Blundell, T.L. Are There Hidden Genes in DNA/RNA Vaccines? Front. Immunol. 2022, 13, 801915. [Google Scholar] [CrossRef] [PubMed]
- Chaudhary, N.; Weissman, D.; Whitehead, K.A. mRNA vaccines for infectious diseases: Principles, delivery and clinical translation. Nat. Rev. Drug Discov. 2021, 20, 817–838. [Google Scholar] [CrossRef]
- Wang, Y.; Zhang, Z.; Luo, J.; Han, X.; Wei, Y.; Wei, X. mRNA vaccine: A potential therapeutic strategy. Mol. Cancer 2021, 20, 33. [Google Scholar] [CrossRef] [PubMed]
- Dolgin, E. The tangled history of mRNA vaccines. Nature 2021, 597, 318–324. [Google Scholar] [CrossRef] [PubMed]
- Schoenmaker, L.; Witzigmann, D.; Kulkarni, J.A.; Verbeke, R.; Kersten, G.; Jiskoot, W.; Crommelin, D.J.A. mRNA-lipid nanoparticle COVID-19 vaccines: Structure and stability. Int. J. Pharm. 2021, 601, 120586. [Google Scholar] [CrossRef] [PubMed]
- De Keersmaecker, B.; Claerhout, S.; Carrasco, J.; Bar, I.; Corthals, J.; Wilgenhof, S.; Neyns, B.; Thielemans, K. TriMix and tumor antigen mRNA electroporated dendritic cell vaccination plus ipilimumab: Link between T-cell activation and clinical responses in advanced melanoma. J. Immunother. Cancer 2020, 8, e000329. [Google Scholar] [CrossRef]
- Wang, S.; Liang, B.; Wang, W.; Li, L.; Feng, N.; Zhao, Y.; Wang, T.; Yan, F.; Yang, S.; Xia, X. Viral vectored vaccines: Design, development, preventive and therapeutic applications in human diseases. Signal Transduct. Target. Ther. 2023, 8, 149. [Google Scholar] [CrossRef]
- Rauch, S.; Jasny, E.; Schmidt, K.E.; Petsch, B. New Vaccine Technologies to Combat Outbreak Situations. Front. Immunol. 2018, 9, 1963. [Google Scholar] [CrossRef]
- Drysdale, S.B.; Barr, R.S.; Rollier, C.S.; Green, C.A.; Pollard, A.J.; Sande, C.J. Priorities for developing respiratory syncytial virus vaccines in different target populations. Sci. Transl. Med. 2020, 12, eaax2466. [Google Scholar] [CrossRef]
- Sun, S.; Liu, Y.; He, C.; Hu, W.; Liu, W.; Huang, X.; Wu, J.; Xie, F.; Chen, C.; Wang, J.; et al. Combining NanoKnife with M1 oncolytic virus enhances anticancer activity in pancreatic cancer. Cancer Lett. 2021, 502, 9–24. [Google Scholar] [CrossRef]
- Soliman, H.; Hogue, D.; Han, H.; Mooney, B.; Costa, R.; Lee, M.C.; Niell, B.; Williams, A.; Chau, A.; Falcon, S.; et al. Oncolytic T-VEC virotherapy plus neoadjuvant chemotherapy in nonmetastatic triple-negative breast cancer: A phase 2 trial. Nat. Med. 2023, 29, 450–457. [Google Scholar] [CrossRef]
- Madan, R.A.; Bilusic, M.; Heery, C.; Schlom, J.; Gulley, J.L. Clinical evaluation of TRICOM vector therapeutic cancer vaccines. Semin. Oncol. 2012, 39, 296–304. [Google Scholar] [CrossRef]
- DeMaria, P.J.; Lee-Wisdom, K.; Donahue, R.N.; Madan, R.A.; Karzai, F.; Schwab, A.; Palena, C.; Jochems, C.; Floudas, C.; Strauss, J.; et al. Phase 1 open-label trial of intravenous administration of MVA-BN-brachyury-TRICOM vaccine in patients with advanced cancer. J. Immunother. Cancer 2021, 9, e003238. [Google Scholar] [CrossRef]
- Gregoriadis, G. Liposomes in Drug Delivery: How It All Happened. Pharmaceutics 2016, 8, 19. [Google Scholar] [CrossRef]
- Huang, T.; Peng, L.; Han, Y.; Wang, D.; He, X.; Wang, J.; Ou, C. Lipid nanoparticle-based mRNA vaccines in cancers: Current advances and future prospects. Front. Immunol. 2022, 13, 922301. [Google Scholar] [CrossRef]
- Chen, J.; Ye, Z.; Huang, C.; Qiu, M.; Song, D.; Li, Y.; Xu, Q. Lipid nanoparticle-mediated lymph node-targeting delivery of mRNA cancer vaccine elicits robust CD8+ T cell response. Proc. Natl. Acad. Sci. USA 2022, 119, e2207841119. [Google Scholar] [CrossRef]
- Poh, A. mRNA Vaccine Slows Melanoma Recurrence. Cancer Discov. 2023, 13, 1278. [Google Scholar] [CrossRef]
- Nagasaka, M.; Potugari, B.; Nguyen, A.; Sukari, A.; Azmi, A.S.; Ou, S.I. KRAS Inhibitors- yes but what next? Direct targeting of KRAS- vaccines, adoptive T cell therapy and beyond. Cancer Treat. Rev. 2021, 101, 102309. [Google Scholar] [CrossRef]
- Gilboa, E. DC-based cancer vaccines. J. Clin. Investig. 2007, 117, 1195–1203. [Google Scholar] [CrossRef]
- Cintolo, J.A.; Datta, J.; Mathew, S.J.; Czerniecki, B.J. Dendritic cell-based vaccines: Barriers and opportunities. Future Oncol. 2012, 8, 1273–1299. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Nagy, N.A.; de Haas, A.M.; Geijtenbeek, T.B.H.; van Ree, R.; Tas, S.W.; van Kooyk, Y.; de Jong, E.C. Therapeutic Liposomal Vaccines for Dendritic Cell Activation or Tolerance. Front. Immunol. 2021, 12, 674048. [Google Scholar] [CrossRef]
- Mastelic-Gavillet, B.; Balint, K.; Boudousquie, C.; Gannon, P.O.; Kandalaft, L.E. Personalized Dendritic Cell Vaccines-Recent Breakthroughs and Encouraging Clinical Results. Front. Immunol. 2019, 10, 766. [Google Scholar] [CrossRef]
- Figlin, R.A.; Tannir, N.M.; Uzzo, R.G.; Tykodi, S.S.; Chen, D.Y.T.; Master, V.; Kapoor, A.; Vaena, D.; Lowrance, W.; Bratslavsky, G.; et al. Results of the ADAPT Phase 3 Study of Rocapuldencel-T in Combination with Sunitinib as First-Line Therapy in Patients with Metastatic Renal Cell Carcinoma. Clin. Cancer Res. 2020, 26, 2327–2336. [Google Scholar] [CrossRef] [PubMed]
- Vogelzang, N.J.; Beer, T.M.; Gerritsen, W.; Oudard, S.; Wiechno, P.; Kukielka-Budny, B.; Samal, V.; Hajek, J.; Feyerabend, S.; Khoo, V.; et al. Efficacy and Safety of Autologous Dendritic Cell-Based Immunotherapy, Docetaxel, and Prednisone vs. Placebo in Patients with Metastatic Castration-Resistant Prostate Cancer: The VIABLE Phase 3 Randomized Clinical Trial. JAMA Oncol. 2022, 8, 546–552. [Google Scholar] [CrossRef]
- Lee, J.B.; Chen, B.; Vasic, D.; Law, A.D.; Zhang, L. Cellular immunotherapy for acute myeloid leukemia: How specific should it be? Blood Rev. 2019, 35, 18–31. [Google Scholar] [CrossRef]
- Batich, K.A.; Mitchell, D.A.; Healy, P.; Herndon, J.E., 2nd; Sampson, J.H. Once, Twice, Three Times a Finding: Reproducibility of Dendritic Cell Vaccine Trials Targeting Cytomegalovirus in Glioblastoma. Clin. Cancer Res. 2020, 26, 5297–5303. [Google Scholar] [CrossRef]
- Sobhani, N.; Scaggiante, B.; Morris, R.; Chai, D.; Catalano, M.; Tardiel-Cyril, D.R.; Neeli, P.; Roviello, G.; Mondani, G.; Li, Y. Therapeutic cancer vaccines: From biological mechanisms and engineering to ongoing clinical trials. Cancer Treat. Rev. 2022, 109, 102429. [Google Scholar] [CrossRef] [PubMed]
- Uddin, M.N.; Roni, M.A. Challenges of Storage and Stability of mRNA-Based COVID-19 Vaccines. Vaccines 2021, 9, 1033. [Google Scholar] [CrossRef]
- Chong, C.; Coukos, G.; Bassani-Sternberg, M. Identification of tumor antigens with immunopeptidomics. Nat. Biotechnol. 2022, 40, 175–188. [Google Scholar] [CrossRef] [PubMed]
- Fan, C.; Qu, H.; Wang, X.; Sobhani, N.; Wang, L.; Liu, S.; Xiong, W.; Zeng, Z.; Li, Y. Cancer/testis antigens: From serology to mRNA cancer vaccine. Semin. Cancer Biol. 2021, 76, 218–231. [Google Scholar] [CrossRef] [PubMed]
- Benn, C.S.; Fisker, A.B.; Rieckmann, A.; Sørup, S.; Aaby, P. Vaccinology: Time to change the paradigm? Lancet Infect. Dis. 2020, 20, e274–e283. [Google Scholar] [CrossRef]
- Huang, X.; Lu, Y.; Guo, M.; Du, S.; Han, N. Recent strategies for nano-based PTT combined with immunotherapy: From a biomaterial point of view. Theranostics 2021, 11, 7546–7569. [Google Scholar] [CrossRef] [PubMed]
- Shemesh, C.S.; Hsu, J.C.; Hosseini, I.; Shen, B.Q.; Rotte, A.; Twomey, P.; Girish, S.; Wu, B. Personalized Cancer Vaccines: Clinical Landscape, Challenges, and Opportunities. Mol.Ther. 2021, 29, 555–570. [Google Scholar] [CrossRef]
- Xu, Z.; Fisher, D.E. mRNA melanoma vaccine revolution spurred by the COVID-19 pandemic. Front. Immunol. 2023, 14, 1155728. [Google Scholar] [CrossRef]
NCT Number | Tumor Type/Target | Phase | Category | Status |
---|---|---|---|---|
NCT03190265 | Pancreatic Cancer | II | Tumor cell | Recruiting |
NCT02451982 | Pancreatic Cancer | I/II | Tumor cell | Recruiting |
NCT03767582 | Advanced PDAC | I/II | Tumor cell | Recruiting |
NCT03376477 | Multiple Myeloma | II | Tumor cell | Recruiting |
NCT03096093 | Neoplasms | I/II | Allogeneic cell | Recruiting |
NCT03970746 | NSCLC | I/II | DC | Recruiting |
NCT03059485 | AML | II | DC | Recruiting |
NCT04523688 | Glioblastoma | II | DC | Not yet recruiting |
NCT03136406 | Pancreatic Cancer/Mutant KRAS | I/II | Virus vector | Active, not recruiting |
NCT03632941 | Breast Cancer/HER2 | II | Virus vector | Recruiting |
NCT03547999 | Metastatic Colorectal Cancer/MVA-BN-CV301 | II | Virus vector | Active, not recruiting |
NCT04747002 | Acute Myeloid Leukemia/DSP-7888 | II | Peptide | Recruiting |
NCT04263051 | Advanced NSCLC/UCPVax | II | Peptide | Recruiting |
NCT03149003 | Glioblastoma/DSP-7888 | III | Peptide | Recruiting |
NCT04206254 | Liver Cancer/gp96 | II/III | Peptide | Not yet recruiting |
NCT04274153 | Human Papilloma Virus/Gardasil9 | IV | Protein | Recruiting |
NCT04090528 | Prostate Cancer, pTVG-HP, pTVG-AR | II | DNA | Recruiting |
NCT03721978, | Cervical cancer/VGX-3100 | III | DNA | Recruiting |
NCT04526899 | Melanoma Stage III-IV/NY-ESO-1, MAGE-A3, Tyrosinase, and TPTE | II | mRNA | Recruiting |
NCT04163094 | Ovarian Cancer W-ova1 | I | mRNA | Recruiting |
NCT03394937 | Resected melanoma (stages IIc, III, and IV)/CD40L, CD70, caTLR4, gp100, MAGE-A3, MAGE-C2, and PRAME | I | mRNA | Recruiting |
NCT02410733 | Melanoma/1 NY-ESO-1, tyrosinase, MAGE-A3, and TPTE | I | mRNA | Recruiting |
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. |
© 2024 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
Katopodi, T.; Petanidis, S.; Grigoriadou, E.; Anestakis, D.; Charalampidis, C.; Chatziprodromidou, I.; Floros, G.; Eskitzis, P.; Zarogoulidis, P.; Koulouris, C.; et al. Immune Specific and Tumor-Dependent mRNA Vaccines for Cancer Immunotherapy: Reprogramming Clinical Translation into Tumor Editing Therapy. Pharmaceutics 2024, 16, 455. https://doi.org/10.3390/pharmaceutics16040455
Katopodi T, Petanidis S, Grigoriadou E, Anestakis D, Charalampidis C, Chatziprodromidou I, Floros G, Eskitzis P, Zarogoulidis P, Koulouris C, et al. Immune Specific and Tumor-Dependent mRNA Vaccines for Cancer Immunotherapy: Reprogramming Clinical Translation into Tumor Editing Therapy. Pharmaceutics. 2024; 16(4):455. https://doi.org/10.3390/pharmaceutics16040455
Chicago/Turabian StyleKatopodi, Theodora, Savvas Petanidis, Eirini Grigoriadou, Doxakis Anestakis, Charalampos Charalampidis, Ioanna Chatziprodromidou, George Floros, Panagiotis Eskitzis, Paul Zarogoulidis, Charilaos Koulouris, and et al. 2024. "Immune Specific and Tumor-Dependent mRNA Vaccines for Cancer Immunotherapy: Reprogramming Clinical Translation into Tumor Editing Therapy" Pharmaceutics 16, no. 4: 455. https://doi.org/10.3390/pharmaceutics16040455
APA StyleKatopodi, T., Petanidis, S., Grigoriadou, E., Anestakis, D., Charalampidis, C., Chatziprodromidou, I., Floros, G., Eskitzis, P., Zarogoulidis, P., Koulouris, C., Sevva, C., Papadopoulos, K., Roulia, P., Mantalovas, S., Dagher, M., Karakousis, A. V., Varsamis, N., Vlassopoulos, K., Theodorou, V., ... Kosmidis, C. (2024). Immune Specific and Tumor-Dependent mRNA Vaccines for Cancer Immunotherapy: Reprogramming Clinical Translation into Tumor Editing Therapy. Pharmaceutics, 16(4), 455. https://doi.org/10.3390/pharmaceutics16040455