Cargando…

Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer

BACKGROUND: Successful targeting of solid tumors such as breast cancer (BC) using chimeric antigen receptor (CAR) T cells has proven challenging, largely attributed to the immunosuppressive tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs) inhibit CAR T cell function and persist...

Descripción completa

Detalles Bibliográficos
Autores principales: Nalawade, Saisha A, Shafer, Paul, Bajgain, Pradip, McKenna, Mary K, Ali, Arushana, Kelly, Lauren, Joubert, Jarrett, Gottschalk, Stephen, Watanabe, Norihiro, Leen, Ann, Parihar, Robin, Vera Valdes, Juan Fernando, Hoyos, Valentina
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8611441/
https://www.ncbi.nlm.nih.gov/pubmed/34815355
http://dx.doi.org/10.1136/jitc-2021-003237
_version_ 1784603296621658112
author Nalawade, Saisha A
Shafer, Paul
Bajgain, Pradip
McKenna, Mary K
Ali, Arushana
Kelly, Lauren
Joubert, Jarrett
Gottschalk, Stephen
Watanabe, Norihiro
Leen, Ann
Parihar, Robin
Vera Valdes, Juan Fernando
Hoyos, Valentina
author_facet Nalawade, Saisha A
Shafer, Paul
Bajgain, Pradip
McKenna, Mary K
Ali, Arushana
Kelly, Lauren
Joubert, Jarrett
Gottschalk, Stephen
Watanabe, Norihiro
Leen, Ann
Parihar, Robin
Vera Valdes, Juan Fernando
Hoyos, Valentina
author_sort Nalawade, Saisha A
collection PubMed
description BACKGROUND: Successful targeting of solid tumors such as breast cancer (BC) using chimeric antigen receptor (CAR) T cells has proven challenging, largely attributed to the immunosuppressive tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs) inhibit CAR T cell function and persistence within the breast TME. To overcome this challenge, we have developed CAR T cells targeting tumor-associated mucin 1 (MUC1) with a novel chimeric costimulatory receptor that targets tumor necrosis factor–related apoptosis-inducing ligand receptor 2 (TR2) expressed on MDSCs. METHODS: The function of the TR2.41BB costimulatory receptor was assessed by exposing non-transduced (NT) and TR2.41BB transduced T cells to recombinant TR2, after which nuclear translocation of NFκB was measured by ELISA and western blot. The cytolytic activity of CAR.MUC1/TR2.41BB T cells was measured in a 5-hour cytotoxicity assay using MUC1+ tumor cells as targets in the presence or absence of MDSCs. In vivo antitumor activity was assessed using MDSC-enriched tumor-bearing mice treated with CAR T cells with or without TR2.41BB. RESULTS: Nuclear translocation of NFκB in response to recombinant TR2 was detected only in TR2.41BB T cells. The presence of MDSCs diminished the cytotoxic potential of CAR.MUC1 T cells against MUC1+ BC cell lines by 25%. However, TR2.41BB expression on CAR.MUC1 T cells induced MDSC apoptosis, thereby restoring the cytotoxic activity of CAR.MUC1 T cells against MUC1+ BC lines. The presence of MDSCs resulted in an approximately twofold increase in tumor growth due to enhanced angiogenesis and fibroblast accumulation compared with mice with tumor alone. Treatment of these MDSC-enriched tumors with CAR.MUC1.TR2.41BB T cells led to superior tumor cell killing and significant reduction in tumor growth (24.54±8.55 mm(3)) compared with CAR.MUC1 (469.79±81.46 mm(3)) or TR2.41BB (434.86±64.25 mm(3)) T cells alone. CAR.MUC1.TR2.41BB T cells also demonstrated improved T cell proliferation and persistence at the tumor site, thereby preventing metastases. We observed similar results using CAR.HER2.TR2.41BB T cells in a HER2+ BC model. CONCLUSIONS: Our findings demonstrate that CAR T cells that coexpress the TR2.4-1BB receptor exhibit superior antitumor potential against breast tumors containing immunosuppressive and tumor promoting MDSCs, resulting in TME remodeling and improved T cell proliferation at the tumor site.
format Online
Article
Text
id pubmed-8611441
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-86114412021-12-10 Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer Nalawade, Saisha A Shafer, Paul Bajgain, Pradip McKenna, Mary K Ali, Arushana Kelly, Lauren Joubert, Jarrett Gottschalk, Stephen Watanabe, Norihiro Leen, Ann Parihar, Robin Vera Valdes, Juan Fernando Hoyos, Valentina J Immunother Cancer Immune Cell Therapies and Immune Cell Engineering BACKGROUND: Successful targeting of solid tumors such as breast cancer (BC) using chimeric antigen receptor (CAR) T cells has proven challenging, largely attributed to the immunosuppressive tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs) inhibit CAR T cell function and persistence within the breast TME. To overcome this challenge, we have developed CAR T cells targeting tumor-associated mucin 1 (MUC1) with a novel chimeric costimulatory receptor that targets tumor necrosis factor–related apoptosis-inducing ligand receptor 2 (TR2) expressed on MDSCs. METHODS: The function of the TR2.41BB costimulatory receptor was assessed by exposing non-transduced (NT) and TR2.41BB transduced T cells to recombinant TR2, after which nuclear translocation of NFκB was measured by ELISA and western blot. The cytolytic activity of CAR.MUC1/TR2.41BB T cells was measured in a 5-hour cytotoxicity assay using MUC1+ tumor cells as targets in the presence or absence of MDSCs. In vivo antitumor activity was assessed using MDSC-enriched tumor-bearing mice treated with CAR T cells with or without TR2.41BB. RESULTS: Nuclear translocation of NFκB in response to recombinant TR2 was detected only in TR2.41BB T cells. The presence of MDSCs diminished the cytotoxic potential of CAR.MUC1 T cells against MUC1+ BC cell lines by 25%. However, TR2.41BB expression on CAR.MUC1 T cells induced MDSC apoptosis, thereby restoring the cytotoxic activity of CAR.MUC1 T cells against MUC1+ BC lines. The presence of MDSCs resulted in an approximately twofold increase in tumor growth due to enhanced angiogenesis and fibroblast accumulation compared with mice with tumor alone. Treatment of these MDSC-enriched tumors with CAR.MUC1.TR2.41BB T cells led to superior tumor cell killing and significant reduction in tumor growth (24.54±8.55 mm(3)) compared with CAR.MUC1 (469.79±81.46 mm(3)) or TR2.41BB (434.86±64.25 mm(3)) T cells alone. CAR.MUC1.TR2.41BB T cells also demonstrated improved T cell proliferation and persistence at the tumor site, thereby preventing metastases. We observed similar results using CAR.HER2.TR2.41BB T cells in a HER2+ BC model. CONCLUSIONS: Our findings demonstrate that CAR T cells that coexpress the TR2.4-1BB receptor exhibit superior antitumor potential against breast tumors containing immunosuppressive and tumor promoting MDSCs, resulting in TME remodeling and improved T cell proliferation at the tumor site. BMJ Publishing Group 2021-11-23 /pmc/articles/PMC8611441/ /pubmed/34815355 http://dx.doi.org/10.1136/jitc-2021-003237 Text en © Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) .
spellingShingle Immune Cell Therapies and Immune Cell Engineering
Nalawade, Saisha A
Shafer, Paul
Bajgain, Pradip
McKenna, Mary K
Ali, Arushana
Kelly, Lauren
Joubert, Jarrett
Gottschalk, Stephen
Watanabe, Norihiro
Leen, Ann
Parihar, Robin
Vera Valdes, Juan Fernando
Hoyos, Valentina
Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer
title Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer
title_full Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer
title_fullStr Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer
title_full_unstemmed Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer
title_short Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer
title_sort selectively targeting myeloid-derived suppressor cells through trail receptor 2 to enhance the efficacy of car t cell therapy for treatment of breast cancer
topic Immune Cell Therapies and Immune Cell Engineering
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8611441/
https://www.ncbi.nlm.nih.gov/pubmed/34815355
http://dx.doi.org/10.1136/jitc-2021-003237
work_keys_str_mv AT nalawadesaishaa selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT shaferpaul selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT bajgainpradip selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT mckennamaryk selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT aliarushana selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT kellylauren selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT joubertjarrett selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT gottschalkstephen selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT watanabenorihiro selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT leenann selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT pariharrobin selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT veravaldesjuanfernando selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer
AT hoyosvalentina selectivelytargetingmyeloidderivedsuppressorcellsthroughtrailreceptor2toenhancetheefficacyofcartcelltherapyfortreatmentofbreastcancer