Cargando…

Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus

BACKGROUND: For programmed cell death protein 1/programmed death-ligand 1 (PD-L1)-positive triple-negative breast cancer (TNBC), the addition of immune checkpoint inhibitors (ICIs) to chemotherapy has been shown to increase the objective response rate (ORR) by only 13.4–16.3%. Thus, examining conver...

Descripción completa

Detalles Bibliográficos
Autores principales: Tang, Jing, Ma, Xuqian, Chen, Xin, Feng, Mingqian, Jin, Xin, Wang, Ke, Wan, Jixi, Zhu, Xianmin, Xie, Rong, Dong, Shuang, Hu, Sheng, Jiang, Hui, Xie, Conghua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9372655/
https://www.ncbi.nlm.nih.gov/pubmed/35965796
http://dx.doi.org/10.21037/atm-22-3423
_version_ 1784767434181312512
author Tang, Jing
Ma, Xuqian
Chen, Xin
Feng, Mingqian
Jin, Xin
Wang, Ke
Wan, Jixi
Zhu, Xianmin
Xie, Rong
Dong, Shuang
Hu, Sheng
Jiang, Hui
Xie, Conghua
author_facet Tang, Jing
Ma, Xuqian
Chen, Xin
Feng, Mingqian
Jin, Xin
Wang, Ke
Wan, Jixi
Zhu, Xianmin
Xie, Rong
Dong, Shuang
Hu, Sheng
Jiang, Hui
Xie, Conghua
author_sort Tang, Jing
collection PubMed
description BACKGROUND: For programmed cell death protein 1/programmed death-ligand 1 (PD-L1)-positive triple-negative breast cancer (TNBC), the addition of immune checkpoint inhibitors (ICIs) to chemotherapy has been shown to increase the objective response rate (ORR) by only 13.4–16.3%. Thus, examining converting tumor immunogenicity and targeting novel pathways may be needed to improve response to immunotherapy. B7-H4 is an emerging target for breast cancer immunotherapy. However, the antitumor effect of B7-H4 is unstable during cell generation and further research is necessary to strengthen the antitumor efficacy of B7-H4. METHODS: To explore the potential of B7-H4-targeted immunotherapy of breast cancer, current study generated four monoclonal antibodies (mAbs) against B7-H4 through immunization of mice followed by phage display technology. T cell-engaged bispecific antibodies and immunotoxins were created based on these mAbs. To evaluate the anti-tumor activity of B7-H4-targeting bispecific antibody and immunotoxin, the anti-tumor efficacy test in vitro and in mice models in vivo were performed. Moreover, to boost the efficacy, oncolytic virus herpes simplex virus type 2 (HSV-2) was combined with the bispecific antibody and tested in mice models. RESULTS: Rigorous in vitro cytotoxicity assay showed that both B7-H4 bispecific antibodies and immunotoxins were toxic to B7-H4 positive cells, with the former being more potent. However, in vivo studies showed that immunotoxin was slightly more effective in suppressing tumor growth. Further, bispecific antibody combined with oncolytic virus HSV-2 revealed a synergistic effect in suppressing tumor growth without causing obvious adverse effects. CONCLUSIONS: Collectively, our findings uncover a novel strategy of combining oncolytic virus HSV-2 with bispecific antibody reinforce antitumor immune response, which warranted further translational investigation.
format Online
Article
Text
id pubmed-9372655
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-93726552022-08-13 Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus Tang, Jing Ma, Xuqian Chen, Xin Feng, Mingqian Jin, Xin Wang, Ke Wan, Jixi Zhu, Xianmin Xie, Rong Dong, Shuang Hu, Sheng Jiang, Hui Xie, Conghua Ann Transl Med Original Article BACKGROUND: For programmed cell death protein 1/programmed death-ligand 1 (PD-L1)-positive triple-negative breast cancer (TNBC), the addition of immune checkpoint inhibitors (ICIs) to chemotherapy has been shown to increase the objective response rate (ORR) by only 13.4–16.3%. Thus, examining converting tumor immunogenicity and targeting novel pathways may be needed to improve response to immunotherapy. B7-H4 is an emerging target for breast cancer immunotherapy. However, the antitumor effect of B7-H4 is unstable during cell generation and further research is necessary to strengthen the antitumor efficacy of B7-H4. METHODS: To explore the potential of B7-H4-targeted immunotherapy of breast cancer, current study generated four monoclonal antibodies (mAbs) against B7-H4 through immunization of mice followed by phage display technology. T cell-engaged bispecific antibodies and immunotoxins were created based on these mAbs. To evaluate the anti-tumor activity of B7-H4-targeting bispecific antibody and immunotoxin, the anti-tumor efficacy test in vitro and in mice models in vivo were performed. Moreover, to boost the efficacy, oncolytic virus herpes simplex virus type 2 (HSV-2) was combined with the bispecific antibody and tested in mice models. RESULTS: Rigorous in vitro cytotoxicity assay showed that both B7-H4 bispecific antibodies and immunotoxins were toxic to B7-H4 positive cells, with the former being more potent. However, in vivo studies showed that immunotoxin was slightly more effective in suppressing tumor growth. Further, bispecific antibody combined with oncolytic virus HSV-2 revealed a synergistic effect in suppressing tumor growth without causing obvious adverse effects. CONCLUSIONS: Collectively, our findings uncover a novel strategy of combining oncolytic virus HSV-2 with bispecific antibody reinforce antitumor immune response, which warranted further translational investigation. AME Publishing Company 2022-07 /pmc/articles/PMC9372655/ /pubmed/35965796 http://dx.doi.org/10.21037/atm-22-3423 Text en 2022 Annals of Translational Medicine. All rights reserved. https://creativecommons.org/licenses/by-nc-nd/4.0/Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0 (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle Original Article
Tang, Jing
Ma, Xuqian
Chen, Xin
Feng, Mingqian
Jin, Xin
Wang, Ke
Wan, Jixi
Zhu, Xianmin
Xie, Rong
Dong, Shuang
Hu, Sheng
Jiang, Hui
Xie, Conghua
Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus
title Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus
title_full Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus
title_fullStr Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus
title_full_unstemmed Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus
title_short Strengthening of antitumor effects in breast cancer from a novel B7-H4- and CD3-targeting bispecific antibody by an oncolytic virus
title_sort strengthening of antitumor effects in breast cancer from a novel b7-h4- and cd3-targeting bispecific antibody by an oncolytic virus
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9372655/
https://www.ncbi.nlm.nih.gov/pubmed/35965796
http://dx.doi.org/10.21037/atm-22-3423
work_keys_str_mv AT tangjing strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT maxuqian strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT chenxin strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT fengmingqian strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT jinxin strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT wangke strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT wanjixi strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT zhuxianmin strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT xierong strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT dongshuang strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT husheng strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT jianghui strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus
AT xieconghua strengtheningofantitumoreffectsinbreastcancerfromanovelb7h4andcd3targetingbispecificantibodybyanoncolyticvirus