CD4+ T-cell epitope-based heterologous prime-boost vaccination potentiates anti-tumor immunity and PD-1/PD-L1 immunotherapy
- PMID: 35580929
- PMCID: PMC9114852
- DOI: 10.1136/jitc-2021-004022
CD4+ T-cell epitope-based heterologous prime-boost vaccination potentiates anti-tumor immunity and PD-1/PD-L1 immunotherapy
Erratum in
-
Correction: CD4+ T-cell epitope-based heterologous prime-boost vaccination potentiates anti-tumor immunity and PD-1/PD-L1 immunotherapy.J Immunother Cancer. 2022 Oct;10(10):e004022corr1. doi: 10.1136/jitc-2021-004022corr1. J Immunother Cancer. 2022. PMID: 36283738 Free PMC article. No abstract available.
Abstract
Background: Antitumor therapeutic vaccines are generally based on antigenic epitopes presented by major histocompatibility complex (MHC-I) molecules to induce tumor-specific CD8+ T cells. Paradoxically, continuous T cell receptor (TCR) stimulation from tumor-derived CD8+ T-cell epitopes can drive the functional exhaustion of tumor-specific CD8+ T cells. Tumor-specific type-I helper CD4+ T (TH1) cells play an important role in the population maintenance and cytotoxic function of exhausted tumor-specific CD8+ T cells in the tumor microenvironment. Nonetheless, whether the vaccination strategy targeting MHC-II-restricted CD4+ T-cell epitopes to induce tumor-specific TH1 responses can confer effective antitumor immunity to restrain tumor growth is not well studied. Here, we developed a heterologous prime-boost vaccination strategy to effectively induce tumor-specific TH1 cells and evaluated its antitumor efficacy and its capacity to potentiate PD-1/PD-L1 immunotherapy.
Methods: Listeria monocytogenes vector and influenza A virus (PR8 strain) vector stably expressing lymphocytic choriomeningitis virus (LCMV) glycoprotein-specific I-Ab-restricted CD4+ T cell epitope (GP61-80) or ovalbumin-specific CD4+ T cell epitope (OVA323-339) were constructed and evaluated their efficacy against mouse models of melanoma and colorectal adenocarcinoma expressing lymphocytic choriomeningitis virus glycoprotein and ovalbumin. The impact of CD4+ T cell epitope-based heterologous prime-boost vaccination was detected by flow-cytometer, single-cell RNA sequencing and single-cell TCR sequencing.
Results: CD4+ T cell epitope-based heterologous prime-boost vaccination efficiently suppressed both mouse melanoma and colorectal adenocarcinoma. This vaccination primarily induced tumor-specific TH1 response, which in turn enhanced the expansion, effector function and clonal breadth of tumor-specific CD8+ T cells. Furthermore, this vaccination strategy synergized PD-L1 blockade mediated tumor suppression. Notably, prime-boost vaccination extended the duration of PD-L1 blockade induced antitumor effects by preventing the re-exhaustion of tumor-specific CD8+ T cells.
Conclusion: CD4+ T cell epitope-based heterologous prime-boost vaccination elicited potent both tumor-specific TH1 and CTL response, leading to the efficient tumor control. This strategy can also potentiate PD-1/PD-L1 immune checkpoint blockade (ICB) against cancer.
Keywords: CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Immunization; Immunotherapy; Vaccination.
© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.
Conflict of interest statement
Competing interests: The authors declare a conflict of interest. A patent associated with a CD4+ T cell epitope-based therapeutic vaccine has been filed (LY and RH).
Figures






Similar articles
-
Efficient control of chronic LCMV infection by a CD4 T cell epitope-based heterologous prime-boost vaccination in a murine model.Cell Mol Immunol. 2018 Sep;15(9):815-826. doi: 10.1038/cmi.2017.3. Epub 2017 Mar 13. Cell Mol Immunol. 2018. PMID: 28287115 Free PMC article.
-
PD-L1 Checkpoint Inhibition Narrows the Antigen-Specific T Cell Receptor Repertoire in Chronic Lymphocytic Choriomeningitis Virus Infection.J Virol. 2020 Aug 31;94(18):e00795-20. doi: 10.1128/JVI.00795-20. Print 2020 Aug 31. J Virol. 2020. PMID: 32641478 Free PMC article.
-
In situ delivery of iPSC-derived dendritic cells with local radiotherapy generates systemic antitumor immunity and potentiates PD-L1 blockade in preclinical poorly immunogenic tumor models.J Immunother Cancer. 2021 May;9(5):e002432. doi: 10.1136/jitc-2021-002432. J Immunother Cancer. 2021. PMID: 34049930 Free PMC article.
-
Reversing T-cell Exhaustion in Cancer: Lessons Learned from PD-1/PD-L1 Immune Checkpoint Blockade.Cancer Immunol Res. 2022 Feb;10(2):146-153. doi: 10.1158/2326-6066.CIR-21-0515. Epub 2021 Dec 22. Cancer Immunol Res. 2022. PMID: 34937730 Review.
-
CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review.J Cell Physiol. 2019 Jun;234(6):8509-8521. doi: 10.1002/jcp.27782. Epub 2018 Nov 22. J Cell Physiol. 2019. PMID: 30520029 Review.
Cited by
-
Targeting tumor metabolism to augment CD8+ T cell anti-tumor immunity.J Pharm Anal. 2025 May;15(5):101150. doi: 10.1016/j.jpha.2024.101150. Epub 2024 Nov 20. J Pharm Anal. 2025. PMID: 40502640 Free PMC article. Review.
-
Viral vector-based therapeutic HPV vaccines.Clin Exp Med. 2024 Aug 28;24(1):199. doi: 10.1007/s10238-024-01470-5. Clin Exp Med. 2024. PMID: 39196444 Free PMC article. Review.
-
Strategies to enhance the therapeutic efficacy of anti-PD-1 antibody, anti-PD-L1 antibody and anti-CTLA-4 antibody in cancer therapy.J Transl Med. 2024 Aug 9;22(1):751. doi: 10.1186/s12967-024-05552-6. J Transl Med. 2024. PMID: 39123227 Free PMC article. Review.
-
Prognostic Value of Liver Kinase B1 (LKB1) in Gastric Cancer-Associated Tumor Microenvironment Immunity.Biomedicines. 2023 Feb 23;11(3):688. doi: 10.3390/biomedicines11030688. Biomedicines. 2023. PMID: 36979667 Free PMC article.
-
Enhanced efficacy of immune checkpoint inhibitors with OK-432 in malignant pleural mesothelioma: A case report.Mol Clin Oncol. 2025 Jul 3;23(3):77. doi: 10.3892/mco.2025.2872. eCollection 2025 Sep. Mol Clin Oncol. 2025. PMID: 40692748 Free PMC article.
References
MeSH terms
Substances
LinkOut - more resources
Full Text Sources
Medical
Research Materials