Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 12/2021

Open Access 01-12-2021 | Gastrointestinal Cancer | Original Article

Combined vaccine-immune-checkpoint inhibition constitutes a promising strategy for treatment of dMMR tumors

Authors: Inken Salewski, Steffen Kuntoff, Andreas Kuemmel, Rico Feldtmann, Stephan B. Felix, Larissa Henze, Christian Junghanss, Claudia Maletzki

Published in: Cancer Immunology, Immunotherapy | Issue 12/2021

Login to get access

Abstract

Background

Mlh1-knock-out-driven mismatch-repair-deficient (dMMR) tumors can be targeted immunologically. By applying therapeutic tumor vaccination, tumor growth is delayed but escape mechanisms evolve, including upregulation of immune-checkpoint molecules (LAG-3, PD-L1). To counteract immune escape, we investigated the therapeutic activity of a combined tumor vaccine-immune-checkpoint inhibitor therapy using α-PD-L1.

Design

In this trial, Mlh1-knock-out mice with established gastrointestinal tumors received single or thrice injections of α-PD-L1 monoclonal antibody clone 6E11 (2.5 mg/kg bw, q2w, i.v.) either alone or in combination with the vaccine. Longitudinal flow cytometry and PET/CT imaging studies were followed by ex vivo functional immunological and gene expression assays.

Results

6E11 monotherapy slightly increased median overall survival (mOS: 6.0 weeks vs. control 4.0 weeks). Increasing the number of injections (n = 3) improved therapy outcome (mOS: 9.2 weeks) and was significantly boosted by combining 6E11 with the vaccine (mOS: 19.4 weeks vs. 10.2 weeks vaccine monotherapy). Accompanying PET/CT imaging confirmed treatment-induced tumor growth control, with the strongest inhibition in the combination group. Three mice (30%) achieved a complete remission and showed long-term survival. Decreased levels of circulating splenic and intratumoral myeloid-derived suppressor cells (MDSC) and decreased numbers of immune-checkpoint-expressing splenic T cells (LAG-3, CTLA-4) accompanied therapeutic effects. Gene expression and protein analysis of residual tumors revealed downregulation of PI3K/Akt/Wnt-and TGF-signaling, leading to T cell infiltration, reduced numbers of macrophages, neutrophils and MDSC.

Conclusions

By successful uncoupling of the PD-1/PD-L1 axis, we provide further evidence for the safe and successful application of immunotherapies to combat dMMR-driven malignancies that warrants further investigation.
Appendix
Available only for authorised users
Literature
1.
go back to reference Cousin S, Seneschal J, Italiano A (2018) Toxicity profiles of immunotherapy. Pharmacol Ther 181:91–100CrossRefPubMed Cousin S, Seneschal J, Italiano A (2018) Toxicity profiles of immunotherapy. Pharmacol Ther 181:91–100CrossRefPubMed
7.
go back to reference Kalim M, Iqbal Khan MS, Zhan J (2020) Programmed cell death ligand-1: a dynamic immune checkpoint in cancer therapy. Chem Biol Drug Des 95:552–566CrossRefPubMed Kalim M, Iqbal Khan MS, Zhan J (2020) Programmed cell death ligand-1: a dynamic immune checkpoint in cancer therapy. Chem Biol Drug Des 95:552–566CrossRefPubMed
15.
18.
go back to reference Lemery S, Keegan P, Pazdur R (2017) First FDA approval agnostic of cancer site—when a biomarker defines the indication. N Engl J Med 377:1409–1412CrossRefPubMed Lemery S, Keegan P, Pazdur R (2017) First FDA approval agnostic of cancer site—when a biomarker defines the indication. N Engl J Med 377:1409–1412CrossRefPubMed
22.
go back to reference Sullivan RJ, Hamid O, Gonzalez R et al (2019) Atezolizumab plus cobimetinib and vemurafenib in BRAF-mutated melanoma patients. Nat Med 25:929–935CrossRefPubMed Sullivan RJ, Hamid O, Gonzalez R et al (2019) Atezolizumab plus cobimetinib and vemurafenib in BRAF-mutated melanoma patients. Nat Med 25:929–935CrossRefPubMed
23.
go back to reference Sahin IH, Akce M, Alese O et al (2019) Immune checkpoint inhibitors for the treatment of MSI-H/MMR-D colorectal cancer and a perspective on resistance mechanisms. Br J Cancer 121:809–818CrossRefPubMedPubMedCentral Sahin IH, Akce M, Alese O et al (2019) Immune checkpoint inhibitors for the treatment of MSI-H/MMR-D colorectal cancer and a perspective on resistance mechanisms. Br J Cancer 121:809–818CrossRefPubMedPubMedCentral
32.
go back to reference Edelmann W, Yang K, Kuraguchi M et al (1999) Tumorigenesis in Mlh1 and Mlh1 / Apc1638N mutant mice. Cancer Res 59:1301–1307PubMed Edelmann W, Yang K, Kuraguchi M et al (1999) Tumorigenesis in Mlh1 and Mlh1 / Apc1638N mutant mice. Cancer Res 59:1301–1307PubMed
33.
go back to reference Chae YK, Anker JF, Bais P, Namburi S (2018) Mutations in DNA repair genes are associated with increased neo-antigen load and activated T cell infiltration in lung adenocarcinoma. Oncotarget 9:7949–7960CrossRefPubMed Chae YK, Anker JF, Bais P, Namburi S (2018) Mutations in DNA repair genes are associated with increased neo-antigen load and activated T cell infiltration in lung adenocarcinoma. Oncotarget 9:7949–7960CrossRefPubMed
34.
go back to reference Chang L, Chang M, Chang HM, Chang F (2018) Microsatellite instability: a predictive biomarker for cancer immunotherapy. Appl Immunohistochem Mol Morphol 26:e15–e21CrossRefPubMed Chang L, Chang M, Chang HM, Chang F (2018) Microsatellite instability: a predictive biomarker for cancer immunotherapy. Appl Immunohistochem Mol Morphol 26:e15–e21CrossRefPubMed
36.
go back to reference Maletzki C, Wiegele L, Nassar I et al (2019) Chemo-immunotherapy improves long- term survival in a preclinical model of MMR-D-related cancer. J Immunother Cancer 7:1–14CrossRef Maletzki C, Wiegele L, Nassar I et al (2019) Chemo-immunotherapy improves long- term survival in a preclinical model of MMR-D-related cancer. J Immunother Cancer 7:1–14CrossRef
49.
go back to reference Shaul ME, Fridlender ZG (2019) Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol 16:601–620CrossRefPubMed Shaul ME, Fridlender ZG (2019) Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol 16:601–620CrossRefPubMed
Metadata
Title
Combined vaccine-immune-checkpoint inhibition constitutes a promising strategy for treatment of dMMR tumors
Authors
Inken Salewski
Steffen Kuntoff
Andreas Kuemmel
Rico Feldtmann
Stephan B. Felix
Larissa Henze
Christian Junghanss
Claudia Maletzki
Publication date
01-12-2021
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 12/2021
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-021-02933-4

Other articles of this Issue 12/2021

Cancer Immunology, Immunotherapy 12/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine