Cargando…

Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) protein is overexpressed on the surface of various epithelial ovarian cancer tissues, mediates the proliferation, differentiation, metastasis, and signal transduction of tumor cells, and thus is a potential cancer therapeutic target. Howeve...

Descripción completa

Detalles Bibliográficos
Autores principales: Zeng, Jinfeng, Zhang, Hui, Guan, Rui
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10331701/
https://www.ncbi.nlm.nih.gov/pubmed/37434690
http://dx.doi.org/10.21037/tcr-23-432
_version_ 1785070297565626368
author Zeng, Jinfeng
Zhang, Hui
Guan, Rui
author_facet Zeng, Jinfeng
Zhang, Hui
Guan, Rui
author_sort Zeng, Jinfeng
collection PubMed
description BACKGROUND: Human epidermal growth factor receptor 2 (HER2) protein is overexpressed on the surface of various epithelial ovarian cancer tissues, mediates the proliferation, differentiation, metastasis, and signal transduction of tumor cells, and thus is a potential cancer therapeutic target. However, its research in ovarian cancer is still limited, and how to quickly obtain a large number of antibodies remains a concern for researchers. METHODS: In this study, we expressed the recombinant anti-HER2 humanized monoclonal antibody (rhHER2-mAb) in human embryonic kidney 293 (HEK293) cells through transient gene expression (TGE) technology by constructing a mammalian cell expression vector. Firstly, the transfection conditions has been optimized, the ratio of light chain (LC) and heavy chain (HC) was optimized in the range of 4:1 to 1:2 and the ration DNA and polyethyleneimine was optimized in the range of 4:1 to 1:1. The antibody was purified by rProtein A affinity chromatography, and its mediated antibody-dependent cellular cytotoxicity (ADCC) was identified by lactate dehydrogenase release assays. The anti-tumor activity of rhHER2-mAb was evaluated in non-obese diabetic/severe combined immunodeficiency mice. RESULTS: The expression of rhHER2-mAb in the HEK293F cells was at the highest level (100.5 mg/L) when the DNA/polyethyleneimine and light-chain/heavy-chain ratios were 1:4 and 1:2, respectively. The half-maximal inhibitory concentration of the ADCC of the antibodies against the SK-OV-3, OVCAR-3, and A-2780 cells were 12.36, 5.43, and 102.90 ng/mL, respectively. The animal experiments with the mice showed that rhHER2-mAb effectively inhibited the growth (P<0.01) of the SK-OV-3 tumors at a dose of 10 mg/kg. CONCLUSIONS: TGE technology allows us to quickly obtain a large number of anti-HER2 antibodies compared to the traditional method of constructing stable cell lines, and its in vitro and in vivo studied shows that our anti-HER2 antibody have higher affinity and better biological activity bioactivity (P<0.01) compared to Herceptin. Our findings provide novel insights into the development and production of future biotechnology-based drugs using the TGE technology of HEK293F.
format Online
Article
Text
id pubmed-10331701
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-103317012023-07-11 Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer Zeng, Jinfeng Zhang, Hui Guan, Rui Transl Cancer Res Original Article BACKGROUND: Human epidermal growth factor receptor 2 (HER2) protein is overexpressed on the surface of various epithelial ovarian cancer tissues, mediates the proliferation, differentiation, metastasis, and signal transduction of tumor cells, and thus is a potential cancer therapeutic target. However, its research in ovarian cancer is still limited, and how to quickly obtain a large number of antibodies remains a concern for researchers. METHODS: In this study, we expressed the recombinant anti-HER2 humanized monoclonal antibody (rhHER2-mAb) in human embryonic kidney 293 (HEK293) cells through transient gene expression (TGE) technology by constructing a mammalian cell expression vector. Firstly, the transfection conditions has been optimized, the ratio of light chain (LC) and heavy chain (HC) was optimized in the range of 4:1 to 1:2 and the ration DNA and polyethyleneimine was optimized in the range of 4:1 to 1:1. The antibody was purified by rProtein A affinity chromatography, and its mediated antibody-dependent cellular cytotoxicity (ADCC) was identified by lactate dehydrogenase release assays. The anti-tumor activity of rhHER2-mAb was evaluated in non-obese diabetic/severe combined immunodeficiency mice. RESULTS: The expression of rhHER2-mAb in the HEK293F cells was at the highest level (100.5 mg/L) when the DNA/polyethyleneimine and light-chain/heavy-chain ratios were 1:4 and 1:2, respectively. The half-maximal inhibitory concentration of the ADCC of the antibodies against the SK-OV-3, OVCAR-3, and A-2780 cells were 12.36, 5.43, and 102.90 ng/mL, respectively. The animal experiments with the mice showed that rhHER2-mAb effectively inhibited the growth (P<0.01) of the SK-OV-3 tumors at a dose of 10 mg/kg. CONCLUSIONS: TGE technology allows us to quickly obtain a large number of anti-HER2 antibodies compared to the traditional method of constructing stable cell lines, and its in vitro and in vivo studied shows that our anti-HER2 antibody have higher affinity and better biological activity bioactivity (P<0.01) compared to Herceptin. Our findings provide novel insights into the development and production of future biotechnology-based drugs using the TGE technology of HEK293F. AME Publishing Company 2023-06-19 2023-06-30 /pmc/articles/PMC10331701/ /pubmed/37434690 http://dx.doi.org/10.21037/tcr-23-432 Text en 2023 Translational Cancer Research. 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
Zeng, Jinfeng
Zhang, Hui
Guan, Rui
Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer
title Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer
title_full Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer
title_fullStr Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer
title_full_unstemmed Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer
title_short Bioactivity of recombinant humanized monoclonal antibody against HER2 in-vivo and in-vitro and its mechanism of action in ovarian cancer
title_sort bioactivity of recombinant humanized monoclonal antibody against her2 in-vivo and in-vitro and its mechanism of action in ovarian cancer
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10331701/
https://www.ncbi.nlm.nih.gov/pubmed/37434690
http://dx.doi.org/10.21037/tcr-23-432
work_keys_str_mv AT zengjinfeng bioactivityofrecombinanthumanizedmonoclonalantibodyagainsther2invivoandinvitroanditsmechanismofactioninovariancancer
AT zhanghui bioactivityofrecombinanthumanizedmonoclonalantibodyagainsther2invivoandinvitroanditsmechanismofactioninovariancancer
AT guanrui bioactivityofrecombinanthumanizedmonoclonalantibodyagainsther2invivoandinvitroanditsmechanismofactioninovariancancer