1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      mRNA vaccine with unmodified uridine induces robust type I interferon-dependent anti-tumor immunity in a melanoma model

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          An mRNA with unmodified nucleosides induces type I interferons (IFN-I) through the stimulation of innate immune sensors. Whether IFN-I induced by mRNA vaccine is crucial for anti-tumor immune response remains to be elucidated. In this study, we investigated the immunogenicity and anti-tumor responses of mRNA encoding tumor antigens with different degrees of N1-methylpseudouridine (m1Ψ) modification in B16 melanoma model. Our results demonstrated that ovalbumin (OVA) encoding mRNA formulated in a lipid nanoparticle (OVA-LNP) induced substantial IFN-I production and the maturation of dendritic cells (DCs) with negative correlation with increasing percentages of m1Ψ modification. In B16-OVA murine melanoma model, unmodified OVA-LNP significantly reduced tumor growth and prolonged survival, compared to OVA-LNP with m1Ψ modification. This robust anti-tumor effect correlated with the increase in intratumoral CD40 + DCs and the frequency of granzyme B +/IFN-γ +/TNF-α + polyfunctional OVA peptide-specific CD8 + T cells. Blocking type I IFN receptor completely reversed the anti-tumor immunity of unmodified mRNA-OVA reflected in a significant decrease in OVA-specific IFN-γ secreting T cells and enrichment of PD-1 + tumor-infiltrating T cells. The robust anti-tumor effect of unmodified OVA-LNP was also observed in the lung metastatic tumor model. Finally, this mRNA vaccine was tested using B16 melanoma neoantigens ( Pbk- Actn4) which resulted in delayed tumor growth. Taken together, our findings demonstrated that an unmodified mRNA vaccine induces IFN-I production or the downstream signaling cascades which plays a crucial role in inducing robust anti-tumor T cell response for controlling tumor growth and metastasis.

          Related collections

          Most cited references56

          • Record: found
          • Abstract: found
          • Article: not found

          mRNA vaccines — a new era in vaccinology

          mRNA vaccines represent a promising alternative to conventional vaccine approaches because of their high potency, capacity for rapid development and potential for low-cost manufacture and safe administration. However, their application has until recently been restricted by the instability and inefficient in vivo delivery of mRNA. Recent technological advances have now largely overcome these issues, and multiple mRNA vaccine platforms against infectious diseases and several types of cancer have demonstrated encouraging results in both animal models and humans. This Review provides a detailed overview of mRNA vaccines and considers future directions and challenges in advancing this promising vaccine platform to widespread therapeutic use.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer

            T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of β2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability.

              In vitro-transcribed mRNAs encoding physiologically important proteins have considerable potential for therapeutic applications. However, in its present form, mRNA is unfeasible for clinical use because of its labile and immunogenic nature. Here, we investigated whether incorporation of naturally modified nucleotides into transcripts would confer enhanced biological properties to mRNA. We found that mRNAs containing pseudouridines have a higher translational capacity than unmodified mRNAs when tested in mammalian cells and lysates or administered intravenously into mice at 0.015-0.15 mg/kg doses. The delivered mRNA and the encoded protein could be detected in the spleen at 1, 4, and 24 hours after the injection, where both products were at significantly higher levels when pseudouridine-containing mRNA was administered. Even at higher doses, only the unmodified mRNA was immunogenic, inducing high serum levels of interferon-alpha (IFN-alpha). These findings indicate that nucleoside modification is an effective approach to enhance stability and translational capacity of mRNA while diminishing its immunogenicity in vivo. Improved properties conferred by pseudouridine make such mRNA a promising tool for both gene replacement and vaccination.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                14 October 2022
                2022
                : 13
                : 983000
                Affiliations
                [1] 1 Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University , Bangkok, Thailand
                [2] 2 Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University , Bangkok, Thailand
                [3] 3 Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA, United States
                [4] 4 Acuitas Therapeutics , Vancouver, BC, Canada
                [5] 5 Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC), Faculty of Medicine, Chulalongkorn University , Bangkok, Thailand
                [6] 6 Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University , Bangkok, Thailand
                [7] 7 Department of Microbiology, Faculty of Science, Chulalongkorn University , Bangkok, Thailand
                Author notes

                Edited by: José Mordoh, IIBBA-CONICET Leloir Institute Foundation, Argentina

                Reviewed by: Koji Yasutomo, Tokushima University, Japan; Rupsa Basu, Humane Genomics, United States

                *Correspondence: Tanapat Palaga, tanapat.p@ 123456chula.ac.th

                This article was submitted to Vaccines and Molecular Therapeutics, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2022.983000
                9614103
                36311701
                2e8c20d6-1573-4253-856b-681a48d86cb3
                Copyright © 2022 Sittplangkoon, Alameh, Weissman, Lin, Tam, Prompetchara and Palaga

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 30 June 2022
                : 26 September 2022
                Page count
                Figures: 8, Tables: 0, Equations: 0, References: 56, Pages: 16, Words: 8366
                Funding
                Funded by: National Research Council of Thailand , doi 10.13039/501100004704;
                Award ID: 811/2563
                Categories
                Immunology
                Original Research

                Immunology
                mrna vaccine,type i interferon,cancer immunotherapy,melanomas,unmodified nucleosides
                Immunology
                mrna vaccine, type i interferon, cancer immunotherapy, melanomas, unmodified nucleosides

                Comments

                Comment on this article