Cargando…

Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo

The PD-1/PD-L1 pathway blockade can generate a good clinical response by reducing immunosuppression and provoking durable antitumor immunity. In addition to antibodies, aptamers can also block the interaction between PD-1 and PD-L1. For the in vivo application, however, free aptamers are usually too...

Descripción completa

Detalles Bibliográficos
Autores principales: An, Yacong, Li, Xundou, Yao, Fengjiao, Duan, Jinhong, Yang, Xian-Da
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8911819/
https://www.ncbi.nlm.nih.gov/pubmed/35268583
http://dx.doi.org/10.3390/molecules27051482
_version_ 1784666932825292800
author An, Yacong
Li, Xundou
Yao, Fengjiao
Duan, Jinhong
Yang, Xian-Da
author_facet An, Yacong
Li, Xundou
Yao, Fengjiao
Duan, Jinhong
Yang, Xian-Da
author_sort An, Yacong
collection PubMed
description The PD-1/PD-L1 pathway blockade can generate a good clinical response by reducing immunosuppression and provoking durable antitumor immunity. In addition to antibodies, aptamers can also block the interaction between PD-1 and PD-L1. For the in vivo application, however, free aptamers are usually too small in size and quickly removed from blood via glomerular filtration. To avoid renal clearance of aptamer, we conjugated the PD-L1 aptamer to albumin to form a larger complex (BSA-Apt) and evaluated whether BSA-Apt would enhance the in vivo antitumor efficacy. The PD-L1 aptamer was thiol-modified and conjugated to the amino group of BSA via a SMCC linker. The average size of BSA-Apt was 11.65 nm, which was above the threshold for renal clearance. Functionally, BSA-Apt retained the capability of the PD-L1 aptamer to bind with PDL1-expressing tumor cells. Moreover, both the free aptamer and BSA-Apt augmented the PBMC-induced antitumor cytotoxicity in vitro. Furthermore, BSA-Apt generated a significantly stronger antitumor efficacy than the free PD-L1 aptamer in vivo without raising systemic toxicity. The results indicate that conjugating the PD-L1 aptamer to albumin may serve as a promising strategy to improve the in vivo functionality of the aptamer and that BSA-Apt may have application potential in cancer immunotherapy.
format Online
Article
Text
id pubmed-8911819
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-89118192022-03-11 Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo An, Yacong Li, Xundou Yao, Fengjiao Duan, Jinhong Yang, Xian-Da Molecules Article The PD-1/PD-L1 pathway blockade can generate a good clinical response by reducing immunosuppression and provoking durable antitumor immunity. In addition to antibodies, aptamers can also block the interaction between PD-1 and PD-L1. For the in vivo application, however, free aptamers are usually too small in size and quickly removed from blood via glomerular filtration. To avoid renal clearance of aptamer, we conjugated the PD-L1 aptamer to albumin to form a larger complex (BSA-Apt) and evaluated whether BSA-Apt would enhance the in vivo antitumor efficacy. The PD-L1 aptamer was thiol-modified and conjugated to the amino group of BSA via a SMCC linker. The average size of BSA-Apt was 11.65 nm, which was above the threshold for renal clearance. Functionally, BSA-Apt retained the capability of the PD-L1 aptamer to bind with PDL1-expressing tumor cells. Moreover, both the free aptamer and BSA-Apt augmented the PBMC-induced antitumor cytotoxicity in vitro. Furthermore, BSA-Apt generated a significantly stronger antitumor efficacy than the free PD-L1 aptamer in vivo without raising systemic toxicity. The results indicate that conjugating the PD-L1 aptamer to albumin may serve as a promising strategy to improve the in vivo functionality of the aptamer and that BSA-Apt may have application potential in cancer immunotherapy. MDPI 2022-02-22 /pmc/articles/PMC8911819/ /pubmed/35268583 http://dx.doi.org/10.3390/molecules27051482 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
An, Yacong
Li, Xundou
Yao, Fengjiao
Duan, Jinhong
Yang, Xian-Da
Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo
title Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo
title_full Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo
title_fullStr Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo
title_full_unstemmed Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo
title_short Novel Complex of PD-L1 Aptamer and Albumin Enhances Antitumor Efficacy In Vivo
title_sort novel complex of pd-l1 aptamer and albumin enhances antitumor efficacy in vivo
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8911819/
https://www.ncbi.nlm.nih.gov/pubmed/35268583
http://dx.doi.org/10.3390/molecules27051482
work_keys_str_mv AT anyacong novelcomplexofpdl1aptamerandalbuminenhancesantitumorefficacyinvivo
AT lixundou novelcomplexofpdl1aptamerandalbuminenhancesantitumorefficacyinvivo
AT yaofengjiao novelcomplexofpdl1aptamerandalbuminenhancesantitumorefficacyinvivo
AT duanjinhong novelcomplexofpdl1aptamerandalbuminenhancesantitumorefficacyinvivo
AT yangxianda novelcomplexofpdl1aptamerandalbuminenhancesantitumorefficacyinvivo