Cargando…

Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion

The arachidonic acid pathway participates in immunosuppression in various types of cancer. Our previous observation detailed that microsomal prostaglandin E2 synthase 1 (mPGES-1), an enzyme downstream of cyclooxygenase 2 (COX-2), limited antitumor immunity in melanoma; in addition, genetic depletion...

Descripción completa

Detalles Bibliográficos
Autores principales: Fukuda, Yasunari, Kim, Sun-Hee, Bustos, Matias A., Cho, Sung-Nam, Roszik, Jason, Burks, Jared K., Kim, Hong, Hoon, Dave S.B., Grimm, Elizabeth A., Ekmekcioglu, Suhendan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Association for Cancer Research 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10389052/
https://www.ncbi.nlm.nih.gov/pubmed/37529399
http://dx.doi.org/10.1158/2767-9764.CRC-23-0210
_version_ 1785082213874794496
author Fukuda, Yasunari
Kim, Sun-Hee
Bustos, Matias A.
Cho, Sung-Nam
Roszik, Jason
Burks, Jared K.
Kim, Hong
Hoon, Dave S.B.
Grimm, Elizabeth A.
Ekmekcioglu, Suhendan
author_facet Fukuda, Yasunari
Kim, Sun-Hee
Bustos, Matias A.
Cho, Sung-Nam
Roszik, Jason
Burks, Jared K.
Kim, Hong
Hoon, Dave S.B.
Grimm, Elizabeth A.
Ekmekcioglu, Suhendan
author_sort Fukuda, Yasunari
collection PubMed
description The arachidonic acid pathway participates in immunosuppression in various types of cancer. Our previous observation detailed that microsomal prostaglandin E2 synthase 1 (mPGES-1), an enzyme downstream of cyclooxygenase 2 (COX-2), limited antitumor immunity in melanoma; in addition, genetic depletion of mPGES-1 specifically enhanced immune checkpoint blockade therapy. The current study set out to distinguish the roles of mPGES-1 from those of COX-2 in tumor immunity and determine the potential of mPGES-1 inhibitors for reinforcing immunotherapy in melanoma. Genetic deletion of mPGES-1 showed different profiles of prostaglandin metabolites from that of COX-2 deletion. In our syngeneic mouse model, mPGES-1–deficient cells exhibited similar tumorigenicity to that of COX-2–deficient cells, despite a lower ability to suppress PGE2 synthesis by mPGES-1 depletion, indicating the presence of factors other than PGE2 that are likely to regulate tumor immunity. RNA-sequencing analysis revealed that mPGES-1 depletion reduced the expressions of collagen-related genes, which have been found to be associated with immunosuppressive signatures. In our mouse model, collagen was reduced in mPGES-1–deficient tumors, and phenotypic analysis of tumor-infiltrating lymphocytes indicated that mPGES-1–deficient tumors had fewer TIM3(+) exhausted CD8(+) T cells compared with COX-2–deficient tumors. CAY10678, an mPGES-1 inhibitor, was equivalent to celecoxib, a selective COX-2 inhibitor, in reinforcing anti-PD-1 treatment. Our study indicates that mPGES-1 inhibitors represent a promising adjuvant for immunotherapies in melanoma by reducing collagen deposition and T-cell exhaustion. SIGNIFICANCE: Collagen is a predominant component of the extracellular matrix that may influence the tumor immune microenvironment for cancer progression. We present here that mPGES-1 has specific roles in regulating tumor immunity, associated with several collagen-related genes and propose that pharmacologic inhibition of mPGES-1 may hold therapeutic promise for improving immune checkpoint–based therapies.
format Online
Article
Text
id pubmed-10389052
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher American Association for Cancer Research
record_format MEDLINE/PubMed
spelling pubmed-103890522023-08-01 Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion Fukuda, Yasunari Kim, Sun-Hee Bustos, Matias A. Cho, Sung-Nam Roszik, Jason Burks, Jared K. Kim, Hong Hoon, Dave S.B. Grimm, Elizabeth A. Ekmekcioglu, Suhendan Cancer Res Commun Research Article The arachidonic acid pathway participates in immunosuppression in various types of cancer. Our previous observation detailed that microsomal prostaglandin E2 synthase 1 (mPGES-1), an enzyme downstream of cyclooxygenase 2 (COX-2), limited antitumor immunity in melanoma; in addition, genetic depletion of mPGES-1 specifically enhanced immune checkpoint blockade therapy. The current study set out to distinguish the roles of mPGES-1 from those of COX-2 in tumor immunity and determine the potential of mPGES-1 inhibitors for reinforcing immunotherapy in melanoma. Genetic deletion of mPGES-1 showed different profiles of prostaglandin metabolites from that of COX-2 deletion. In our syngeneic mouse model, mPGES-1–deficient cells exhibited similar tumorigenicity to that of COX-2–deficient cells, despite a lower ability to suppress PGE2 synthesis by mPGES-1 depletion, indicating the presence of factors other than PGE2 that are likely to regulate tumor immunity. RNA-sequencing analysis revealed that mPGES-1 depletion reduced the expressions of collagen-related genes, which have been found to be associated with immunosuppressive signatures. In our mouse model, collagen was reduced in mPGES-1–deficient tumors, and phenotypic analysis of tumor-infiltrating lymphocytes indicated that mPGES-1–deficient tumors had fewer TIM3(+) exhausted CD8(+) T cells compared with COX-2–deficient tumors. CAY10678, an mPGES-1 inhibitor, was equivalent to celecoxib, a selective COX-2 inhibitor, in reinforcing anti-PD-1 treatment. Our study indicates that mPGES-1 inhibitors represent a promising adjuvant for immunotherapies in melanoma by reducing collagen deposition and T-cell exhaustion. SIGNIFICANCE: Collagen is a predominant component of the extracellular matrix that may influence the tumor immune microenvironment for cancer progression. We present here that mPGES-1 has specific roles in regulating tumor immunity, associated with several collagen-related genes and propose that pharmacologic inhibition of mPGES-1 may hold therapeutic promise for improving immune checkpoint–based therapies. American Association for Cancer Research 2023-07-31 /pmc/articles/PMC10389052/ /pubmed/37529399 http://dx.doi.org/10.1158/2767-9764.CRC-23-0210 Text en © 2023 The Authors; Published by the American Association for Cancer Research https://creativecommons.org/licenses/by/4.0/This open access article is distributed under the Creative Commons Attribution 4.0 International (CC BY 4.0) license.
spellingShingle Research Article
Fukuda, Yasunari
Kim, Sun-Hee
Bustos, Matias A.
Cho, Sung-Nam
Roszik, Jason
Burks, Jared K.
Kim, Hong
Hoon, Dave S.B.
Grimm, Elizabeth A.
Ekmekcioglu, Suhendan
Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion
title Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion
title_full Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion
title_fullStr Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion
title_full_unstemmed Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion
title_short Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-cell Exhaustion
title_sort inhibition of microsomal prostaglandin e2 synthase reduces collagen deposition in melanoma tumors and may improve immunotherapy efficacy by reducing t-cell exhaustion
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10389052/
https://www.ncbi.nlm.nih.gov/pubmed/37529399
http://dx.doi.org/10.1158/2767-9764.CRC-23-0210
work_keys_str_mv AT fukudayasunari inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT kimsunhee inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT bustosmatiasa inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT chosungnam inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT roszikjason inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT burksjaredk inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT kimhong inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT hoondavesb inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT grimmelizabetha inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion
AT ekmekcioglusuhendan inhibitionofmicrosomalprostaglandine2synthasereducescollagendepositioninmelanomatumorsandmayimproveimmunotherapyefficacybyreducingtcellexhaustion