Cargando…

Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer

BACKGROUND: Agents blocking programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) have been approved for triple-negative breast cancer (TNBC). However, the response rate of anti-PD-1/PD-L1 is still unsatisfactory, partly due to immunosuppressive factors such as transforming growth f...

Descripción completa

Detalles Bibliográficos
Autores principales: Yi, Ming, Wu, Yuze, Niu, Mengke, Zhu, Shuangli, Zhang, Jing, Yan, Yongxiang, Zhou, Pengfei, Dai, Zhijun, Wu, Kongming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9723957/
https://www.ncbi.nlm.nih.gov/pubmed/36460337
http://dx.doi.org/10.1136/jitc-2022-005543
_version_ 1784844305626562560
author Yi, Ming
Wu, Yuze
Niu, Mengke
Zhu, Shuangli
Zhang, Jing
Yan, Yongxiang
Zhou, Pengfei
Dai, Zhijun
Wu, Kongming
author_facet Yi, Ming
Wu, Yuze
Niu, Mengke
Zhu, Shuangli
Zhang, Jing
Yan, Yongxiang
Zhou, Pengfei
Dai, Zhijun
Wu, Kongming
author_sort Yi, Ming
collection PubMed
description BACKGROUND: Agents blocking programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) have been approved for triple-negative breast cancer (TNBC). However, the response rate of anti-PD-1/PD-L1 is still unsatisfactory, partly due to immunosuppressive factors such as transforming growth factor-beta (TGF-β). In our previous pilot study, the bispecific antibody targeting TGF-β and murine PD-L1 (termed YM101) showed potent antitumor effect. In this work, we constructed a bispecific antibody targeting TGF-β and human PD-L1 (termed BiTP) and explored the antitumor effect of BiTP in TNBC. METHODS: BiTP was developed using Check-BODY(TM) bispecific platform. The binding affinity of BiTP was measured by surface plasmon resonance, ELISA, and flow cytometry. The bioactivity was assessed by Smad and NFAT luciferase reporter assays, immunofluorescence, western blotting, and superantigen stimulation assays. The antitumor activity of BiTP was explored in humanized epithelial-mesenchymal transition-6-hPDL1 and 4T1-hPDL1 murine TNBC models. Immunohistochemical staining, flow cytometry, and bulk RNA-seq were used to investigate the effect of BiTP on immune cell infiltration. RESULTS: BiTP exhibited high binding affinity to dual targets. In vitro experiments verified that BiTP effectively counteracted TGF-β-Smad and PD-L1-PD-1-NFAT signaling. In vivo animal experiments demonstrated that BiTP had superior antitumor activity relative to anti-PD-L1 and anti-TGF-β monotherapy. Mechanistically, BiTP decreased collagen deposition, enhanced CD8(+) T cell penetration, and increased tumor-infiltrating lymphocytes. This improved tumor microenvironment contributed to the potent antitumor activity of BiTP. CONCLUSION: BiTP retains parent antibodies’ binding affinity and bioactivity, with superior antitumor activity to parent antibodies in TNBC. Our data suggest that BiTP might be a promising agent for TNBC treatment.
format Online
Article
Text
id pubmed-9723957
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-97239572022-12-07 Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer Yi, Ming Wu, Yuze Niu, Mengke Zhu, Shuangli Zhang, Jing Yan, Yongxiang Zhou, Pengfei Dai, Zhijun Wu, Kongming J Immunother Cancer Clinical/Translational Cancer Immunotherapy BACKGROUND: Agents blocking programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) have been approved for triple-negative breast cancer (TNBC). However, the response rate of anti-PD-1/PD-L1 is still unsatisfactory, partly due to immunosuppressive factors such as transforming growth factor-beta (TGF-β). In our previous pilot study, the bispecific antibody targeting TGF-β and murine PD-L1 (termed YM101) showed potent antitumor effect. In this work, we constructed a bispecific antibody targeting TGF-β and human PD-L1 (termed BiTP) and explored the antitumor effect of BiTP in TNBC. METHODS: BiTP was developed using Check-BODY(TM) bispecific platform. The binding affinity of BiTP was measured by surface plasmon resonance, ELISA, and flow cytometry. The bioactivity was assessed by Smad and NFAT luciferase reporter assays, immunofluorescence, western blotting, and superantigen stimulation assays. The antitumor activity of BiTP was explored in humanized epithelial-mesenchymal transition-6-hPDL1 and 4T1-hPDL1 murine TNBC models. Immunohistochemical staining, flow cytometry, and bulk RNA-seq were used to investigate the effect of BiTP on immune cell infiltration. RESULTS: BiTP exhibited high binding affinity to dual targets. In vitro experiments verified that BiTP effectively counteracted TGF-β-Smad and PD-L1-PD-1-NFAT signaling. In vivo animal experiments demonstrated that BiTP had superior antitumor activity relative to anti-PD-L1 and anti-TGF-β monotherapy. Mechanistically, BiTP decreased collagen deposition, enhanced CD8(+) T cell penetration, and increased tumor-infiltrating lymphocytes. This improved tumor microenvironment contributed to the potent antitumor activity of BiTP. CONCLUSION: BiTP retains parent antibodies’ binding affinity and bioactivity, with superior antitumor activity to parent antibodies in TNBC. Our data suggest that BiTP might be a promising agent for TNBC treatment. BMJ Publishing Group 2022-12-02 /pmc/articles/PMC9723957/ /pubmed/36460337 http://dx.doi.org/10.1136/jitc-2022-005543 Text en © 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. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) .
spellingShingle Clinical/Translational Cancer Immunotherapy
Yi, Ming
Wu, Yuze
Niu, Mengke
Zhu, Shuangli
Zhang, Jing
Yan, Yongxiang
Zhou, Pengfei
Dai, Zhijun
Wu, Kongming
Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer
title Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer
title_full Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer
title_fullStr Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer
title_full_unstemmed Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer
title_short Anti-TGF-β/PD-L1 bispecific antibody promotes T cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer
title_sort anti-tgf-β/pd-l1 bispecific antibody promotes t cell infiltration and exhibits enhanced antitumor activity in triple-negative breast cancer
topic Clinical/Translational Cancer Immunotherapy
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9723957/
https://www.ncbi.nlm.nih.gov/pubmed/36460337
http://dx.doi.org/10.1136/jitc-2022-005543
work_keys_str_mv AT yiming antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT wuyuze antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT niumengke antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT zhushuangli antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT zhangjing antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT yanyongxiang antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT zhoupengfei antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT daizhijun antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer
AT wukongming antitgfbpdl1bispecificantibodypromotestcellinfiltrationandexhibitsenhancedantitumoractivityintriplenegativebreastcancer