Cargando…

Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy

Epacadostat (EPA), the most advanced IDO1 inhibitor, in combination with PD-1 checkpoint inhibitor, has failed in a recent Phase III clinical trial for treating metastatic melanoma. Here we report an EPA nanovesicle therapeutic platform (Epacasome) based on chemically attaching EPA to sphingomyelin...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Zhiren, Li, Wenpan, Jiang, Yanhao, Tran, Tuyen Ba, Cordova, Leyla Estrella, Chung, Jinha, Kim, Minhyeok, Wondrak, Georg, Erdrich, Jennifer, Lu, Jianqin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10636136/
https://www.ncbi.nlm.nih.gov/pubmed/37945606
http://dx.doi.org/10.1038/s41467-023-43079-4
_version_ 1785133148227502080
author Wang, Zhiren
Li, Wenpan
Jiang, Yanhao
Tran, Tuyen Ba
Cordova, Leyla Estrella
Chung, Jinha
Kim, Minhyeok
Wondrak, Georg
Erdrich, Jennifer
Lu, Jianqin
author_facet Wang, Zhiren
Li, Wenpan
Jiang, Yanhao
Tran, Tuyen Ba
Cordova, Leyla Estrella
Chung, Jinha
Kim, Minhyeok
Wondrak, Georg
Erdrich, Jennifer
Lu, Jianqin
author_sort Wang, Zhiren
collection PubMed
description Epacadostat (EPA), the most advanced IDO1 inhibitor, in combination with PD-1 checkpoint inhibitor, has failed in a recent Phase III clinical trial for treating metastatic melanoma. Here we report an EPA nanovesicle therapeutic platform (Epacasome) based on chemically attaching EPA to sphingomyelin via an oxime-ester bond highly responsive to hydrolase cleavage. Via clathrin-mediated endocytosis, Epacasome displays higher cellular uptake and enhances IDO1 inhibition and T cell proliferation compared to free EPA. Epacasome shows improved pharmacokinetics and tumour accumulation with efficient intratumoural drug release and deep tumour penetration. Additionally, it outperforms free EPA for anticancer efficacy, potentiating PD-1 blockade with boosted cytotoxic T lymphocytes (CTLs) and reduced regulatory T cells and myeloid-derived suppressor cells responses in a B16-F10 melanoma model in female mice. By co-encapsulating immunogenic dacarbazine, Epacasome further enhances anti-tumor effects and immune responses through the upregulation of NKG2D-mediated CTLs and natural killer cells responses particularly when combined with the PD-1 inhibitor in the late-stage metastatic B16-F10-Luc2 model in female mice. Furthermore, this combination prevents tumour recurrence and prolongs mouse survival in a clinically relevant, post-surgical melanoma model in female mice. Epacasome demonstrates potential to synergize with PD-1 blockade for improved response to melanoma immunotherapy.
format Online
Article
Text
id pubmed-10636136
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-106361362023-11-11 Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy Wang, Zhiren Li, Wenpan Jiang, Yanhao Tran, Tuyen Ba Cordova, Leyla Estrella Chung, Jinha Kim, Minhyeok Wondrak, Georg Erdrich, Jennifer Lu, Jianqin Nat Commun Article Epacadostat (EPA), the most advanced IDO1 inhibitor, in combination with PD-1 checkpoint inhibitor, has failed in a recent Phase III clinical trial for treating metastatic melanoma. Here we report an EPA nanovesicle therapeutic platform (Epacasome) based on chemically attaching EPA to sphingomyelin via an oxime-ester bond highly responsive to hydrolase cleavage. Via clathrin-mediated endocytosis, Epacasome displays higher cellular uptake and enhances IDO1 inhibition and T cell proliferation compared to free EPA. Epacasome shows improved pharmacokinetics and tumour accumulation with efficient intratumoural drug release and deep tumour penetration. Additionally, it outperforms free EPA for anticancer efficacy, potentiating PD-1 blockade with boosted cytotoxic T lymphocytes (CTLs) and reduced regulatory T cells and myeloid-derived suppressor cells responses in a B16-F10 melanoma model in female mice. By co-encapsulating immunogenic dacarbazine, Epacasome further enhances anti-tumor effects and immune responses through the upregulation of NKG2D-mediated CTLs and natural killer cells responses particularly when combined with the PD-1 inhibitor in the late-stage metastatic B16-F10-Luc2 model in female mice. Furthermore, this combination prevents tumour recurrence and prolongs mouse survival in a clinically relevant, post-surgical melanoma model in female mice. Epacasome demonstrates potential to synergize with PD-1 blockade for improved response to melanoma immunotherapy. Nature Publishing Group UK 2023-11-09 /pmc/articles/PMC10636136/ /pubmed/37945606 http://dx.doi.org/10.1038/s41467-023-43079-4 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Wang, Zhiren
Li, Wenpan
Jiang, Yanhao
Tran, Tuyen Ba
Cordova, Leyla Estrella
Chung, Jinha
Kim, Minhyeok
Wondrak, Georg
Erdrich, Jennifer
Lu, Jianqin
Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy
title Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy
title_full Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy
title_fullStr Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy
title_full_unstemmed Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy
title_short Sphingomyelin-derived nanovesicles for the delivery of the IDO1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy
title_sort sphingomyelin-derived nanovesicles for the delivery of the ido1 inhibitor epacadostat enhance metastatic and post-surgical melanoma immunotherapy
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10636136/
https://www.ncbi.nlm.nih.gov/pubmed/37945606
http://dx.doi.org/10.1038/s41467-023-43079-4
work_keys_str_mv AT wangzhiren sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT liwenpan sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT jiangyanhao sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT trantuyenba sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT cordovaleylaestrella sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT chungjinha sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT kimminhyeok sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT wondrakgeorg sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT erdrichjennifer sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy
AT lujianqin sphingomyelinderivednanovesiclesforthedeliveryoftheido1inhibitorepacadostatenhancemetastaticandpostsurgicalmelanomaimmunotherapy