Cargando…

A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy

BACKGROUND: PD-1/PD-L1 immune checkpoint inhibitors are currently the most commonly utilized agents in clinical practice, which elicit an immunostimulatory response to combat malignancies. However, all these inhibitors are currently administered via injection using antibody-based therapies, while th...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Dan, Wang, Jingmei, You, Weiming, Ma, Fang, Sun, Qi, She, Junjun, He, Wangxiao, Yang, Guang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10388715/
https://www.ncbi.nlm.nih.gov/pubmed/37529049
http://dx.doi.org/10.3389/fimmu.2023.1228581
_version_ 1785082180213407744
author Liu, Dan
Wang, Jingmei
You, Weiming
Ma, Fang
Sun, Qi
She, Junjun
He, Wangxiao
Yang, Guang
author_facet Liu, Dan
Wang, Jingmei
You, Weiming
Ma, Fang
Sun, Qi
She, Junjun
He, Wangxiao
Yang, Guang
author_sort Liu, Dan
collection PubMed
description BACKGROUND: PD-1/PD-L1 immune checkpoint inhibitors are currently the most commonly utilized agents in clinical practice, which elicit an immunostimulatory response to combat malignancies. However, all these inhibitors are currently administered via injection using antibody-based therapies, while there is a growing need for oral alternatives. METHODS: This study has developed and synthesized exosome-wrapped gold–peptide nanocomplexes with low immunogenicity, which can target PD-L1 and activate antitumor immunity in vivo through oral absorption. The (Super)PDL1(exo) was characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS), Fourier transform infrared (FTIR), X-ray photoelectron spectroscopy (XPS), and gel silver staining. The transmembrane ability of (Super)PDL1(exo) was evaluated by flow cytometry and immunofluorescence. Cell viability was determined using the Cell Counting Kit-8 (CCK-8) assay. ELISA experiments were conducted to detect serum and tissue inflammatory factors, as well as serum biochemical indicators. Tissue sections were stained with H&E for the evaluation of the safety of (Super)PDL1(exo). An MC38 colon cancer model was established in immunocompetent C56BL/6 mice to evaluate the effects of (Super)PDL1(exo) on tumor growth in vivo. Immunohistochemistry (IHC) staining was performed to detect cytotoxicity factors such as perforin and granzymes. RESULTS: First, (Super)PDL1 was successfully synthesized, and milk exosome membranes were encapsulated through ultrasound, repeated freeze–thaw cycles, and extrusion, resulting in the synthesis of (Super)PDL1(exo). Multiple characterization results confirmed the successful synthesis of (Super)PDL1(exo) nanoparticles. Furthermore, our data demonstrated that (Super)PDL1(exo) exhibited excellent colloidal stability and superior cell transmembrane ability. In vitro and in vivo experiments revealed that (Super)PDL1(exo) did not cause damage to multiple systemic organs, demonstrating its good biocompatibility. Finally, in the MC38 colon cancer mouse model, it was discovered that (Super)PDL1(exo) could inhibit the progression of colon cancer, and this tumor-suppressive effect was mediated through the activation of tumor-specific cytotoxic T lymphocyte (CTL)-related immune responses. CONCLUSION: This study has successfully designed and synthesized an oral nanotherapeutic, (Super)PDL1(exo), which demonstrates small particle size, excellent colloidal stability, transmembrane ability in tumor cells, and biocompatibility. In vivo experiments have shown that it effectively activates T-cell immunity and exerts antitumor effects.
format Online
Article
Text
id pubmed-10388715
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-103887152023-08-01 A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy Liu, Dan Wang, Jingmei You, Weiming Ma, Fang Sun, Qi She, Junjun He, Wangxiao Yang, Guang Front Immunol Immunology BACKGROUND: PD-1/PD-L1 immune checkpoint inhibitors are currently the most commonly utilized agents in clinical practice, which elicit an immunostimulatory response to combat malignancies. However, all these inhibitors are currently administered via injection using antibody-based therapies, while there is a growing need for oral alternatives. METHODS: This study has developed and synthesized exosome-wrapped gold–peptide nanocomplexes with low immunogenicity, which can target PD-L1 and activate antitumor immunity in vivo through oral absorption. The (Super)PDL1(exo) was characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS), Fourier transform infrared (FTIR), X-ray photoelectron spectroscopy (XPS), and gel silver staining. The transmembrane ability of (Super)PDL1(exo) was evaluated by flow cytometry and immunofluorescence. Cell viability was determined using the Cell Counting Kit-8 (CCK-8) assay. ELISA experiments were conducted to detect serum and tissue inflammatory factors, as well as serum biochemical indicators. Tissue sections were stained with H&E for the evaluation of the safety of (Super)PDL1(exo). An MC38 colon cancer model was established in immunocompetent C56BL/6 mice to evaluate the effects of (Super)PDL1(exo) on tumor growth in vivo. Immunohistochemistry (IHC) staining was performed to detect cytotoxicity factors such as perforin and granzymes. RESULTS: First, (Super)PDL1 was successfully synthesized, and milk exosome membranes were encapsulated through ultrasound, repeated freeze–thaw cycles, and extrusion, resulting in the synthesis of (Super)PDL1(exo). Multiple characterization results confirmed the successful synthesis of (Super)PDL1(exo) nanoparticles. Furthermore, our data demonstrated that (Super)PDL1(exo) exhibited excellent colloidal stability and superior cell transmembrane ability. In vitro and in vivo experiments revealed that (Super)PDL1(exo) did not cause damage to multiple systemic organs, demonstrating its good biocompatibility. Finally, in the MC38 colon cancer mouse model, it was discovered that (Super)PDL1(exo) could inhibit the progression of colon cancer, and this tumor-suppressive effect was mediated through the activation of tumor-specific cytotoxic T lymphocyte (CTL)-related immune responses. CONCLUSION: This study has successfully designed and synthesized an oral nanotherapeutic, (Super)PDL1(exo), which demonstrates small particle size, excellent colloidal stability, transmembrane ability in tumor cells, and biocompatibility. In vivo experiments have shown that it effectively activates T-cell immunity and exerts antitumor effects. Frontiers Media S.A. 2023-07-17 /pmc/articles/PMC10388715/ /pubmed/37529049 http://dx.doi.org/10.3389/fimmu.2023.1228581 Text en Copyright © 2023 Liu, Wang, You, Ma, Sun, She, He and Yang https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Liu, Dan
Wang, Jingmei
You, Weiming
Ma, Fang
Sun, Qi
She, Junjun
He, Wangxiao
Yang, Guang
A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy
title A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy
title_full A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy
title_fullStr A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy
title_full_unstemmed A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy
title_short A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy
title_sort d-peptide-based oral nanotherapeutic modulates the pd-1/pd-l1 interaction for tumor immunotherapy
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10388715/
https://www.ncbi.nlm.nih.gov/pubmed/37529049
http://dx.doi.org/10.3389/fimmu.2023.1228581
work_keys_str_mv AT liudan adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT wangjingmei adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT youweiming adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT mafang adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT sunqi adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT shejunjun adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT hewangxiao adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT yangguang adpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT liudan dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT wangjingmei dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT youweiming dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT mafang dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT sunqi dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT shejunjun dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT hewangxiao dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy
AT yangguang dpeptidebasedoralnanotherapeuticmodulatesthepd1pdl1interactionfortumorimmunotherapy