Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 Mar 3;29(3):1186-1198.
doi: 10.1016/j.ymthe.2020.11.027. Epub 2020 Dec 3.

Self-Replicating RNAs Drive Protective Anti-tumor T Cell Responses to Neoantigen Vaccine Targets in a Combinatorial Approach

Affiliations

Self-Replicating RNAs Drive Protective Anti-tumor T Cell Responses to Neoantigen Vaccine Targets in a Combinatorial Approach

Christian J Maine et al. Mol Ther. .

Abstract

Historically poor clinical results of tumor vaccines have been attributed to weakly immunogenic antigen targets, limited specificity, and vaccine platforms that fail to induce high-quality polyfunctional T cells, central to mediating cellular immunity. We show here that the combination of antigen selection, construct design, and a robust vaccine platform based on the Synthetically Modified Alpha Replicon RNA Technology (SMARRT), a self-replicating RNA, leads to control of tumor growth in mice. Therapeutic immunization with SMARRT replicon-based vaccines expressing tumor-specific neoantigens or tumor-associated antigen were able to generate polyfunctional CD4+ and CD8+ T cell responses in mice. Additionally, checkpoint inhibitors, or co-administration of cytokine also expressed from the SMARRT platform, synergized to enhance responses further. Lastly, SMARRT-based immunization of non-human primates was able to elicit high-quality T cell responses, demonstrating translatability and clinical feasibility of synthetic replicon technology for therapeutic oncology vaccines.

Keywords: RNA; T cell; immuno-oncology; neoantigen; replicon; self-replicating; synthetic; tumor; vaccine.

PubMed Disclaimer

Conflict of interest statement

All authors affiliated with Synthetic Genomics Inc. declare no competing interests. A.D.G. is a senior officer and a majority shareholder, and L.M. is an employee of EpiVax, Inc., a privately owned immunoinformatics and vaccine design company. These authors acknowledge that there is a potential conflict of interest related to their relationship with EpiVax and attest that the work contained in this research report is free of any bias that might be associated with the commercial goals of the company. G.B. was previously a senior officer of EpiVax Therapeutics, Inc., and G.R. is currently an employee of EpiVax Therapeutics, Inc., a precision immunotherapy company and subsidiary of EpiVax, Inc. These authors acknowledge that there is a potential conflict of interest related to their relationship with EpiVax Therapeutics and attest that the work contained in this research report is free of any bias that might be associated with the commercial goals of the company.

Figures

None
Graphical abstract
Figure 1
Figure 1
Ancer-Designed Polytope Neoantigen Constructs Expressed in the SMARRT Platform Generate Polyfunctional CD4 and CD8 T Cell Responses upon Vaccination (A) Schematic showing polytope design considerations following neoantigen identification. SMARRT replicons expressing neoantigen cassettes designed with Ancer were injected into BALB/c mice at a single dose of 10 μg, and spleens were removed 14 days later and restimulated with a peptide pool containing the top 50 Ancer predicted neoantigens from CT26. T cell function was measured by IFN-γ ELISpot (B), and intracellular cytokine staining is shown for selected replicons (C). Graphs show mean with standard deviation, n = 5 mice per group. Statistical testing was carried out with ordinary one-way ANOVA. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗∗p < 0.0001.
Figure 2
Figure 2
SMARRT.Ancer Primes a Limited Number of Dominant T Cell Epitopes SMARRT.Ancer (containing the top 20 CT26 neoantigens) was used to immunize BALB/c mice with varying prime/boost interval lengths. Splenocytes were analyzed by intracellular cytokine staining (on the indicated days post-final injection) by restimulating individually with all 20 neoantigens encoded in the cassette. (A) and (B) show the percentage of IFN-γ+ CD8 and CD4 T cells specific for peptides stimulating a response above mock (data not shown). (C) and (D) show the polyfunctionality of CD8 and CD4 T cells in response to the dominant neoantigen 20 and 4, respectively. Graphs show mean with standard deviation, n = 5 mice per group. Statistical analysis was performed using one-way ANOVA with Tukeys multiple comparison test. For (C) and (D), the triple functional subsets were compared. p < 0.05; ∗∗p < 0.01; ∗∗∗∗p < 0.0001
Figure 3
Figure 3
SMARRT Can Prime Polyfunctional T Cell Responses Specific to the Shared Neoantigen KRAS G12V HLA-A∗1101 transgenic mice were vaccinated with SMARRT replicons encoding an epitope from either WT or G12V mutant KRAS and boosted on days 21 and 41. Spleens were analyzed 7 days post final boost by intracellular cytokine staining (A) and IFN-γ ELISpot (B) following restimulation ex vivo with the indicated peptides. Graphs show mean with standard deviation, n = 3 mice per group. Statistical analysis was performed using Mann-Whitney U Test, panel B compared double cytokine positive T cells. p<0.05; ∗∗∗∗p<0.0001.
Figure 4
Figure 4
Therapeutic Vaccination with SMARRT.Ancer Inhibits Tumor Growth and Enhances Survival Mice were implanted with 3 × 105 CT26 cells s.c. at day 0. SMARRT was injected i.m. on days 3 and 17. Tumor growth (A) and overall survival (B) were measured (n = 18/group). In a separate study, mice (n = 6/group) were injected with CT26 and SMARRT constructs as described above; additionally, some groups received anti-PD-1 blocking mAbs on days 8, 11, 15, and 18. (C) Shows tumor growth until day 23. Tumors were excised on day 23 and T cells were analyzed by flow cytometry for cytokine production following ex vivo peptide re-stimulation. (D) Shows CD8 T cells and (E) shows CD4 T cells. Tumor growth curves show median tumor volume, and statistical testing was done with two-way ANOVA. Survival curve statistical testing was done using log-rank (Mantel Cox) test. Bar graphs show mean with standard deviation. ∗p < 0.05; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001.
Figure 5
Figure 5
SMARRT Can Be Used to Express Cytokines, Resulting in Epitope Spreading to Weakly Immunogenic Tumor-Associated Antigens SMARRT expressing hIL-7/15 fusion, mIL-7/15 fusion, mIL-7, mIL-15, mGM-CSF, and IL-12 were used to transfect BHK cells. (A) Supernatants were harvested 24 h post-electroporation and cytokine concentrations were measured using ELISA. SMARRT.Trp2 was used to immunize C57BL/6 mice on days 0 and 14. Spleens were harvested on day 21 and restimulated with the dominant CD8 epitope TRP2180–188. (B and C) T cell function was measured by IFN-γ ELISpot (B) and intracellular cytokine staining (C). (D) Mice were immunized as described above with SMARRT.Trp2; in addition, mice were immunized with SMARRT.IL-12 at prime only, as indicated in the figure. The graph shows intracellular cytokine staining following stimulation with an overlapping peptide pool of TRP2 (Sub.) or TRP2180–188 (Dom.). Graphs in (B)–(D) show mean with standard deviation, n = 5 mice per group. Statistical testing for (B) and (C) was done using Mann-Whitney U test. Statistical testing in (D) was done using ordinary one-way ANOVA to compare the total IFN-γ+ CD8 T cells, ∗p < 0.05; ∗∗p < 0.01.
Figure 6
Figure 6
SMARRT Primes Polyfunctional T Cell Responses in Non-human Primates SMARRT replicons expressing HA from the H1N1 or H5N1 strains of influenza virus were used to immunize rhesus macaques on day 0 and 56 at the indicated doses. Saline immunization was used as a control. The animals were bled on day 77 and PBMCs were re-stimulated with peptide pools encoding either H1 or H5 HA sequences. (A) and (B) show activated (OX40+ CD25+) CD8 and CD4 T cells found in the PBMCs as measured by flow cytometry (solid circles, LNP formulated 100 μg; open circles, unformulated 100 μg; squares, formulated 10 μg; triangles, formulated 1 μg). (C) and (D) show cytokine production of CD8 and CD4 T cells measured by flow cytometry. Each bar/symbol corresponds to one animal. Statistical testing was carried out with one-tailed Mann-Whitney U test, ∗p < 0.05.

References

    1. Hollingsworth R.E., Jansen K. Turning the corner on therapeutic cancer vaccines. npj. Vaccines (Basel) 2019;4:1–10. - PMC - PubMed
    1. Castle J.C., Kreiter S., Diekmann J., Löwer M., van de Roemer N., de Graaf J., Selmi A., Diken M., Boegel S., Paret C. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72:1081–1091. - PubMed
    1. Guo Y., Lei K., Tang L. Neoantigen Vaccine Delivery for Personalized Anticancer Immunotherapy. Front. Immunol. 2018;9:1499. - PMC - PubMed
    1. De Groot A.S., Moise L., Terry F., Gutierrez A.H., Hindocha P., Richard G., Hoft D.F., Ross T.M., Noe A.R., Takahashi Y. Better Epitope Discovery, Precision Immune Engineering, and Accelerated Vaccine Design Using Immunoinformatics Tools. Front. Immunol. 2020;11:442. - PMC - PubMed
    1. Chen D.S., Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10. - PubMed