CD4+ T-cell epitope-based heterologous prime-boost vaccination potentiates anti-tumor immunity and PD-1/PD-L1 immunotherapy

© 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..

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.

Errataetall:

ErratumIn: J Immunother Cancer. 2022 Oct;10(10):. - PMID 36283738

Medienart:

E-Artikel

Erscheinungsjahr:

2022

Erschienen:

2022

Enthalten in:

Zur Gesamtaufnahme - volume:10

Enthalten in:

Journal for immunotherapy of cancer - 10(2022), 5 vom: 26. Mai

Sprache:

Englisch

Beteiligte Personen:

Xiao, Minglu [VerfasserIn]
Xie, Luoyingzi [VerfasserIn]
Cao, Guoshuai [VerfasserIn]
Lei, Shun [VerfasserIn]
Wang, Pengcheng [VerfasserIn]
Wei, Zhengping [VerfasserIn]
Luo, Yuan [VerfasserIn]
Fang, Jingyi [VerfasserIn]
Yang, Xingxing [VerfasserIn]
Huang, Qizhao [VerfasserIn]
Xu, Lifan [VerfasserIn]
Guo, Junyi [VerfasserIn]
Wen, Shuqiong [VerfasserIn]
Wang, Zhiming [VerfasserIn]
Wu, Qing [VerfasserIn]
Tang, Jianfang [VerfasserIn]
Wang, Lisha [VerfasserIn]
Chen, Xiangyu [VerfasserIn]
Chen, Cheng [VerfasserIn]
Zhang, Yanyan [VerfasserIn]
Yao, Wei [VerfasserIn]
Ye, Jianqiang [VerfasserIn]
He, Ran [VerfasserIn]
Huang, Jun [VerfasserIn]
Ye, Lilin [VerfasserIn]

Links:

Volltext

Themen:

9006-59-1
B7-H1 Antigen
CD4-Positive T-Lymphocytes
CD8-Positive T-Lymphocytes
Epitopes, T-Lymphocyte
Glycoproteins
Immune Checkpoint Inhibitors
Immunization
Immunotherapy
Journal Article
Ovalbumin
Programmed Cell Death 1 Receptor
Receptors, Antigen, T-Cell
Vaccination

Anmerkungen:

Date Completed 20.05.2022

Date Revised 25.10.2022

published: Print

ErratumIn: J Immunother Cancer. 2022 Oct;10(10):. - PMID 36283738

Citation Status MEDLINE

doi:

10.1136/jitc-2021-004022

funding:

Förderinstitution / Projekttitel:

PPN (Katalog-ID):

NLM34103732X