Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests

Background Cellular immunotherapies using autologous tumor-infiltrating lymphocytes (TIL) can induce durable regression of epithelial cancers in selected patients with treatment-refractory metastatic disease. As the genetic engineering of T cells with tumor-reactive T-cell receptors (TCRs) comes to...

Full description

Bibliographic Details
Main Authors: Steven A Rosenberg, Paul Robbins, Biman Paria, Jared J Gartner, Sri Krishna, Satyajit Ray, Rami Yossef, Victoria Hill, Frank J Lowery, Praveen D Chatani, Neilesh B Parikh, Kyle J Hitscherich, Maria Florentin, Alakesh Bera, Maria Parkhust
Format: Article
Language:English
Published: BMJ Publishing Group 2023-05-01
Series:Journal for ImmunoTherapy of Cancer
Online Access:https://jitc.bmj.com/content/11/5/e006264.full
_version_ 1797814585598672896
author Steven A Rosenberg
Paul Robbins
Biman Paria
Jared J Gartner
Sri Krishna
Satyajit Ray
Rami Yossef
Victoria Hill
Frank J Lowery
Praveen D Chatani
Neilesh B Parikh
Kyle J Hitscherich
Maria Florentin
Alakesh Bera
Maria Parkhust
author_facet Steven A Rosenberg
Paul Robbins
Biman Paria
Jared J Gartner
Sri Krishna
Satyajit Ray
Rami Yossef
Victoria Hill
Frank J Lowery
Praveen D Chatani
Neilesh B Parikh
Kyle J Hitscherich
Maria Florentin
Alakesh Bera
Maria Parkhust
author_sort Steven A Rosenberg
collection DOAJ
description Background Cellular immunotherapies using autologous tumor-infiltrating lymphocytes (TIL) can induce durable regression of epithelial cancers in selected patients with treatment-refractory metastatic disease. As the genetic engineering of T cells with tumor-reactive T-cell receptors (TCRs) comes to the forefront of clinical investigation, the rapid, scalable, and cost-effective detection of patient-specific neoantigen-reactive TIL remains a top priority.Methods We analyzed the single-cell transcriptomic states of 31 neoantigen-specific T-cell clonotypes to identify cell surface dysfunction markers that best identified the metastatic transcriptional states enriched with antitumor TIL. We developed an efficient method to capture neoantigen-reactive TCRs directly from resected human tumors based on cell surface co-expression of CD39, programmed cell death protein-1, and TIGIT dysfunction markers (CD8+ TILTP).Results TILTP TCR isolation achieved a high degree of correlation with single-cell transcriptomic signatures that identify neoantigen-reactive TCRs, making it a cost-effective strategy using widely available resources. Reconstruction of additional TILTP TCRs from tumors identified known and novel antitumor TCRs, showing that at least 39.5% of TILTP TCRs are neoantigen-reactive or tumor-reactive. Despite their substantial enrichment for neoantigen-reactive TCR clonotypes, clonal dynamics of 24 unique antitumor TILTP clonotypes from four patients indicated that most in vitro expanded TILTP populations failed to demonstrate neoantigen reactivity, either by loss of neoantigen-reactive clones during TIL expansion, or through functional impairment during cognate neoantigen recognition.Conclusions While direct usage of in vitro-expanded CD8+ TILTP as a source for cellular therapy might be precluded by profound TIL dysfunction, isolating TILTP represents a streamlined effective approach to rapidly identify neoantigen-reactive TCRs to design engineered cellular immunotherapies against cancer.
first_indexed 2024-03-13T08:09:50Z
format Article
id doaj.art-cbbdaffbcb90479ea2c903c47528272b
institution Directory Open Access Journal
issn 2051-1426
language English
last_indexed 2024-03-13T08:09:50Z
publishDate 2023-05-01
publisher BMJ Publishing Group
record_format Article
series Journal for ImmunoTherapy of Cancer
spelling doaj.art-cbbdaffbcb90479ea2c903c47528272b2023-05-31T23:30:06ZengBMJ Publishing GroupJournal for ImmunoTherapy of Cancer2051-14262023-05-0111510.1136/jitc-2022-006264Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digestsSteven A Rosenberg0Paul Robbins1Biman Paria2Jared J Gartner3Sri Krishna4Satyajit Ray5Rami Yossef6Victoria Hill7Frank J Lowery8Praveen D Chatani9Neilesh B Parikh10Kyle J Hitscherich11Maria Florentin12Alakesh Bera13Maria Parkhust14Surgery Branch, National Cancer Institute, Bethesda, Maryland, USASurgery Branch, National Cancer Institute, Bethesda, Maryland, USAProgram Coordination and Referral Branch, National Cancer Institute, Bethesda, Maryland, USASurgery Branch, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USADepartment of paediatrics, Royal Hospital for Children, Glasgow, UKSurgery Branch, National Cancer Institute, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USASurgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USABackground Cellular immunotherapies using autologous tumor-infiltrating lymphocytes (TIL) can induce durable regression of epithelial cancers in selected patients with treatment-refractory metastatic disease. As the genetic engineering of T cells with tumor-reactive T-cell receptors (TCRs) comes to the forefront of clinical investigation, the rapid, scalable, and cost-effective detection of patient-specific neoantigen-reactive TIL remains a top priority.Methods We analyzed the single-cell transcriptomic states of 31 neoantigen-specific T-cell clonotypes to identify cell surface dysfunction markers that best identified the metastatic transcriptional states enriched with antitumor TIL. We developed an efficient method to capture neoantigen-reactive TCRs directly from resected human tumors based on cell surface co-expression of CD39, programmed cell death protein-1, and TIGIT dysfunction markers (CD8+ TILTP).Results TILTP TCR isolation achieved a high degree of correlation with single-cell transcriptomic signatures that identify neoantigen-reactive TCRs, making it a cost-effective strategy using widely available resources. Reconstruction of additional TILTP TCRs from tumors identified known and novel antitumor TCRs, showing that at least 39.5% of TILTP TCRs are neoantigen-reactive or tumor-reactive. Despite their substantial enrichment for neoantigen-reactive TCR clonotypes, clonal dynamics of 24 unique antitumor TILTP clonotypes from four patients indicated that most in vitro expanded TILTP populations failed to demonstrate neoantigen reactivity, either by loss of neoantigen-reactive clones during TIL expansion, or through functional impairment during cognate neoantigen recognition.Conclusions While direct usage of in vitro-expanded CD8+ TILTP as a source for cellular therapy might be precluded by profound TIL dysfunction, isolating TILTP represents a streamlined effective approach to rapidly identify neoantigen-reactive TCRs to design engineered cellular immunotherapies against cancer.https://jitc.bmj.com/content/11/5/e006264.full
spellingShingle Steven A Rosenberg
Paul Robbins
Biman Paria
Jared J Gartner
Sri Krishna
Satyajit Ray
Rami Yossef
Victoria Hill
Frank J Lowery
Praveen D Chatani
Neilesh B Parikh
Kyle J Hitscherich
Maria Florentin
Alakesh Bera
Maria Parkhust
Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests
Journal for ImmunoTherapy of Cancer
title Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests
title_full Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests
title_fullStr Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests
title_full_unstemmed Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests
title_short Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests
title_sort cell surface marker based capture of neoantigen reactive cd8 t cell receptors from metastatic tumor digests
url https://jitc.bmj.com/content/11/5/e006264.full
work_keys_str_mv AT stevenarosenberg cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT paulrobbins cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT bimanparia cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT jaredjgartner cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT srikrishna cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT satyajitray cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT ramiyossef cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT victoriahill cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT frankjlowery cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT praveendchatani cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT neileshbparikh cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT kylejhitscherich cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT mariaflorentin cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT alakeshbera cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests
AT mariaparkhust cellsurfacemarkerbasedcaptureofneoantigenreactivecd8tcellreceptorsfrommetastatictumordigests