Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4

Abstract Background: Antibodies and derivative drugs targeting immune checkpoints have been approved for the treatment of several malignancies, but there are fewer responses in patients with pancreatic cancer. Here, we designed a nanobody molecule with bi-targeting on PD-L1 and CXCR4, as both target...

Full description

Bibliographic Details
Main Authors: Shuai Hao, Shuyi Xu, Liangzhu Li, Yaxian Li, Meiqi Zhao, Junsheng Chen, Shunying Zhu, Yueqing Xie, Hua Jiang, Jianwei Zhu, Mingyuan Wu
Format: Article
Language:English
Published: BMC 2022-10-01
Series:BMC Cancer
Subjects:
Online Access:https://doi.org/10.1186/s12885-022-10165-7
_version_ 1797991613522247680
author Shuai Hao
Shuyi Xu
Liangzhu Li
Yaxian Li
Meiqi Zhao
Junsheng Chen
Shunying Zhu
Yueqing Xie
Hua Jiang
Jianwei Zhu
Mingyuan Wu
author_facet Shuai Hao
Shuyi Xu
Liangzhu Li
Yaxian Li
Meiqi Zhao
Junsheng Chen
Shunying Zhu
Yueqing Xie
Hua Jiang
Jianwei Zhu
Mingyuan Wu
author_sort Shuai Hao
collection DOAJ
description Abstract Background: Antibodies and derivative drugs targeting immune checkpoints have been approved for the treatment of several malignancies, but there are fewer responses in patients with pancreatic cancer. Here, we designed a nanobody molecule with bi-targeting on PD-L1 and CXCR4, as both targets are overexpressed in many cancer cells and play important roles in tumorigenesis. We characterized the biochemical and anti-tumour activities of the bispecific nanobodies in vitro and in vivo. Methods: A nanobody molecule was designed and constructed. The nanobody sequences targeting PD-L1 and CXCR4 were linked by the (G4S)3 flexible peptide to construct the anti-PD-L1/CXCR4 bispecific nanobody. The bispecific nanobody was expressed in E. coli cells and purified by affinity chromatography. The purified nanobody was biochemically characterized by mass spectrometry, Western blotting and flow cytometry to confirm the molecule and its association with both PD-L1 and CXCR4. The biological function of the nanobody and its anti-tumour effects were examined by an in vitro tumour cell-killing assay and in vivo tumour inhibition in mouse xenograft models. Results: A novel anti-PD-L1/CXCR4 bispecific nanobody was designed, constructed and characterized. The molecule specifically bound to two targets on the surface of human cancer cells and inhibited CXCL12-induced Jurkat cell migration. The bispecific nanobody increased the level of IFN-γ secreted by T-cell activation. The cytotoxicity of human peripheral blood mononuclear cells (hPBMCs) against pancreatic cancer cells was enhanced by the molecule in combination with IL-2. In a human pancreatic cancer xenograft model, the anti-PD-L1/CXCR4 nanobody markedly inhibited tumour growth and was superior to the combo-treatment by anti-PD-L1 nanobody and anti-CXCR4 nanobody or treatment with atezolizumab as a positive control. Immunofluorescence and immunohistochemical staining of xenograft tumours showed that the anti-tumour effects were associated with the inhibition of angiogenesis and the infiltration of immune cells. Conclusion: These results clearly revealed that the anti-PD-L1/CXCR4 bispecific nanobody exerted anti-tumour efficacy in vitro and inhibited tumour growth in vivo. This agent can be further developed as a therapeutic reagent to treat human pancreatic cancer by simultaneously blocking two critical targets.
first_indexed 2024-04-11T08:55:00Z
format Article
id doaj.art-3e00bfdc6861413c8cde585309b0cf39
institution Directory Open Access Journal
issn 1471-2407
language English
last_indexed 2024-04-11T08:55:00Z
publishDate 2022-10-01
publisher BMC
record_format Article
series BMC Cancer
spelling doaj.art-3e00bfdc6861413c8cde585309b0cf392022-12-22T04:33:19ZengBMCBMC Cancer1471-24072022-10-0122111210.1186/s12885-022-10165-7Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4Shuai Hao0Shuyi Xu1Liangzhu Li2Yaxian Li3Meiqi Zhao4Junsheng Chen5Shunying Zhu6Yueqing Xie7Hua Jiang8Jianwei Zhu9Mingyuan Wu10Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityEngineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityEngineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityEngineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityEngineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityEngineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityInstitute of Translational Medicine, Shanghai Jiao Tong UniversityJecho Laboratories, IncJecho Laboratories, IncEngineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityEngineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong UniversityAbstract Background: Antibodies and derivative drugs targeting immune checkpoints have been approved for the treatment of several malignancies, but there are fewer responses in patients with pancreatic cancer. Here, we designed a nanobody molecule with bi-targeting on PD-L1 and CXCR4, as both targets are overexpressed in many cancer cells and play important roles in tumorigenesis. We characterized the biochemical and anti-tumour activities of the bispecific nanobodies in vitro and in vivo. Methods: A nanobody molecule was designed and constructed. The nanobody sequences targeting PD-L1 and CXCR4 were linked by the (G4S)3 flexible peptide to construct the anti-PD-L1/CXCR4 bispecific nanobody. The bispecific nanobody was expressed in E. coli cells and purified by affinity chromatography. The purified nanobody was biochemically characterized by mass spectrometry, Western blotting and flow cytometry to confirm the molecule and its association with both PD-L1 and CXCR4. The biological function of the nanobody and its anti-tumour effects were examined by an in vitro tumour cell-killing assay and in vivo tumour inhibition in mouse xenograft models. Results: A novel anti-PD-L1/CXCR4 bispecific nanobody was designed, constructed and characterized. The molecule specifically bound to two targets on the surface of human cancer cells and inhibited CXCL12-induced Jurkat cell migration. The bispecific nanobody increased the level of IFN-γ secreted by T-cell activation. The cytotoxicity of human peripheral blood mononuclear cells (hPBMCs) against pancreatic cancer cells was enhanced by the molecule in combination with IL-2. In a human pancreatic cancer xenograft model, the anti-PD-L1/CXCR4 nanobody markedly inhibited tumour growth and was superior to the combo-treatment by anti-PD-L1 nanobody and anti-CXCR4 nanobody or treatment with atezolizumab as a positive control. Immunofluorescence and immunohistochemical staining of xenograft tumours showed that the anti-tumour effects were associated with the inhibition of angiogenesis and the infiltration of immune cells. Conclusion: These results clearly revealed that the anti-PD-L1/CXCR4 bispecific nanobody exerted anti-tumour efficacy in vitro and inhibited tumour growth in vivo. This agent can be further developed as a therapeutic reagent to treat human pancreatic cancer by simultaneously blocking two critical targets.https://doi.org/10.1186/s12885-022-10165-7PD-L1CXCR4AntibodyBispecific nanobodyImmunotherapyPancreatic cancer
spellingShingle Shuai Hao
Shuyi Xu
Liangzhu Li
Yaxian Li
Meiqi Zhao
Junsheng Chen
Shunying Zhu
Yueqing Xie
Hua Jiang
Jianwei Zhu
Mingyuan Wu
Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4
BMC Cancer
PD-L1
CXCR4
Antibody
Bispecific nanobody
Immunotherapy
Pancreatic cancer
title Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4
title_full Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4
title_fullStr Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4
title_full_unstemmed Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4
title_short Tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting PD-L1 and CXCR4
title_sort tumour inhibitory activity on pancreatic cancer by bispecific nanobody targeting pd l1 and cxcr4
topic PD-L1
CXCR4
Antibody
Bispecific nanobody
Immunotherapy
Pancreatic cancer
url https://doi.org/10.1186/s12885-022-10165-7
work_keys_str_mv AT shuaihao tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT shuyixu tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT liangzhuli tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT yaxianli tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT meiqizhao tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT junshengchen tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT shunyingzhu tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT yueqingxie tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT huajiang tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT jianweizhu tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4
AT mingyuanwu tumourinhibitoryactivityonpancreaticcancerbybispecificnanobodytargetingpdl1andcxcr4