Cargando…

BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control

BACKGROUND: Cancer immunotherapies can produce complete therapeutic responses, however, outcomes in ovarian cancer (OC) are modest. While adoptive T-cell transfer (ACT) has been evaluated in OC, durable effects are rare. Poor therapeutic efficacy is likely multifactorial, stemming from limited antig...

Descripción completa

Detalles Bibliográficos
Autores principales: McGray, A J Robert, Chiello, Jessie L, Tsuji, Takemasa, Long, Mark, Maraszek, Kathryn, Gaulin, Nicole, Rosario, Spencer R, Hess, Suzanne M, Abrams, Scott I, Kozbor, Danuta, Odunsi, Kunle, Zsiros, Emese
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10314690/
https://www.ncbi.nlm.nih.gov/pubmed/37647218
http://dx.doi.org/10.1136/jitc-2023-006863
_version_ 1785067363914219520
author McGray, A J Robert
Chiello, Jessie L
Tsuji, Takemasa
Long, Mark
Maraszek, Kathryn
Gaulin, Nicole
Rosario, Spencer R
Hess, Suzanne M
Abrams, Scott I
Kozbor, Danuta
Odunsi, Kunle
Zsiros, Emese
author_facet McGray, A J Robert
Chiello, Jessie L
Tsuji, Takemasa
Long, Mark
Maraszek, Kathryn
Gaulin, Nicole
Rosario, Spencer R
Hess, Suzanne M
Abrams, Scott I
Kozbor, Danuta
Odunsi, Kunle
Zsiros, Emese
author_sort McGray, A J Robert
collection PubMed
description BACKGROUND: Cancer immunotherapies can produce complete therapeutic responses, however, outcomes in ovarian cancer (OC) are modest. While adoptive T-cell transfer (ACT) has been evaluated in OC, durable effects are rare. Poor therapeutic efficacy is likely multifactorial, stemming from limited antigen recognition, insufficient tumor targeting due to a suppressive tumor microenvironment (TME), and limited intratumoral accumulation/persistence of infused T cells. Importantly, host T cells infiltrate tumors, and ACT approaches that leverage endogenous tumor-infiltrating T cells for antitumor immunity could effectively magnify therapeutic responses. METHODS: Using retroviral transduction, we have generated T cells that secrete a folate receptor alpha (FRα)-directed bispecific T-cell engager (FR-B T cells), a tumor antigen commonly overexpressed in OC and other tumor types. The antitumor activity and therapeutic efficacy of FR-B T cells was assessed using FRα+ cancer cell lines, OC patient samples, and preclinical tumor models with accompanying mechanistic studies. Different cytokine stimulation of T cells (interleukin (IL)-2+IL-7 vs IL-2+IL-15) during FR-B T cell production and the resulting impact on therapeutic outcome following ACT was also assessed. RESULTS: FR-B T cells efficiently lysed FRα+ cell lines, targeted FRα+ OC patient tumor cells, and were found to engage and activate patient T cells present in the TME through secretion of T cell engagers. Additionally, FR-B T cell therapy was effective in an immunocompetent in vivo OC model, with response duration dependent on both endogenous T cells and FR-B T cell persistence. IL-2/IL-15 preconditioning prior to ACT produced less differentiated FR-B T cells and enhanced therapeutic efficacy, with mechanistic studies revealing preferential accumulation of TCF-1+CD39−CD69− stem-like CD8+ FR B T cells in the peritoneal cavity over solid tumors. CONCLUSIONS: These findings highlight the therapeutic potential of FR-B T cells in OC and suggest FR-B T cells can persist in extratumoral spaces while actively directing antitumor immunity. As the therapeutic activity of infused T cell therapies in solid tumor indications is often limited by poor intratumoral accumulation of transferred T cells, engager-secreting T cells that can effectively leverage endogenous immunity may have distinct mechanistic advantages for enhancing therapeutic responses rates.
format Online
Article
Text
id pubmed-10314690
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-103146902023-07-02 BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control McGray, A J Robert Chiello, Jessie L Tsuji, Takemasa Long, Mark Maraszek, Kathryn Gaulin, Nicole Rosario, Spencer R Hess, Suzanne M Abrams, Scott I Kozbor, Danuta Odunsi, Kunle Zsiros, Emese J Immunother Cancer Immune Cell Therapies and Immune Cell Engineering BACKGROUND: Cancer immunotherapies can produce complete therapeutic responses, however, outcomes in ovarian cancer (OC) are modest. While adoptive T-cell transfer (ACT) has been evaluated in OC, durable effects are rare. Poor therapeutic efficacy is likely multifactorial, stemming from limited antigen recognition, insufficient tumor targeting due to a suppressive tumor microenvironment (TME), and limited intratumoral accumulation/persistence of infused T cells. Importantly, host T cells infiltrate tumors, and ACT approaches that leverage endogenous tumor-infiltrating T cells for antitumor immunity could effectively magnify therapeutic responses. METHODS: Using retroviral transduction, we have generated T cells that secrete a folate receptor alpha (FRα)-directed bispecific T-cell engager (FR-B T cells), a tumor antigen commonly overexpressed in OC and other tumor types. The antitumor activity and therapeutic efficacy of FR-B T cells was assessed using FRα+ cancer cell lines, OC patient samples, and preclinical tumor models with accompanying mechanistic studies. Different cytokine stimulation of T cells (interleukin (IL)-2+IL-7 vs IL-2+IL-15) during FR-B T cell production and the resulting impact on therapeutic outcome following ACT was also assessed. RESULTS: FR-B T cells efficiently lysed FRα+ cell lines, targeted FRα+ OC patient tumor cells, and were found to engage and activate patient T cells present in the TME through secretion of T cell engagers. Additionally, FR-B T cell therapy was effective in an immunocompetent in vivo OC model, with response duration dependent on both endogenous T cells and FR-B T cell persistence. IL-2/IL-15 preconditioning prior to ACT produced less differentiated FR-B T cells and enhanced therapeutic efficacy, with mechanistic studies revealing preferential accumulation of TCF-1+CD39−CD69− stem-like CD8+ FR B T cells in the peritoneal cavity over solid tumors. CONCLUSIONS: These findings highlight the therapeutic potential of FR-B T cells in OC and suggest FR-B T cells can persist in extratumoral spaces while actively directing antitumor immunity. As the therapeutic activity of infused T cell therapies in solid tumor indications is often limited by poor intratumoral accumulation of transferred T cells, engager-secreting T cells that can effectively leverage endogenous immunity may have distinct mechanistic advantages for enhancing therapeutic responses rates. BMJ Publishing Group 2023-06-23 /pmc/articles/PMC10314690/ /pubmed/37647218 http://dx.doi.org/10.1136/jitc-2023-006863 Text en © Author(s) (or their employer(s)) 2023. 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
McGray, A J Robert
Chiello, Jessie L
Tsuji, Takemasa
Long, Mark
Maraszek, Kathryn
Gaulin, Nicole
Rosario, Spencer R
Hess, Suzanne M
Abrams, Scott I
Kozbor, Danuta
Odunsi, Kunle
Zsiros, Emese
BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control
title BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control
title_full BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control
title_fullStr BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control
title_full_unstemmed BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control
title_short BiTE secretion by adoptively transferred stem-like T cells improves FRα+ ovarian cancer control
title_sort bite secretion by adoptively transferred stem-like t cells improves frα+ ovarian cancer control
topic Immune Cell Therapies and Immune Cell Engineering
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10314690/
https://www.ncbi.nlm.nih.gov/pubmed/37647218
http://dx.doi.org/10.1136/jitc-2023-006863
work_keys_str_mv AT mcgrayajrobert bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT chiellojessiel bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT tsujitakemasa bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT longmark bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT maraszekkathryn bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT gaulinnicole bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT rosariospencerr bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT hesssuzannem bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT abramsscotti bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT kozbordanuta bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT odunsikunle bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol
AT zsirosemese bitesecretionbyadoptivelytransferredstemliketcellsimprovesfraovariancancercontrol