SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint

SLAMF6 is a homotypic receptor of the Ig-superfamily whose exact role in immune modulation has remained elusive. Its constitutive expression on resting and activated T cells precludes it from being a bona fide exhaustion marker. By breeding Pmel-1 mice with SLAMF6 -/- mice, we generated donors for T...

Full description

Bibliographic Details
Main Authors: Emma Hajaj, Galit Eisenberg, Shiri Klein, Shoshana Frankenburg, Sharon Merims, Inna Ben David, Thomas Eisenhaure, Sarah E Henrickson, Alexandra Chloé Villani, Nir Hacohen, Nathalie Abudi, Rinat Abramovich, Jonathan E Cohen, Tamar Peretz, Andre Veillette, Michal Lotem
Format: Article
Language:English
Published: eLife Sciences Publications Ltd 2020-03-01
Series:eLife
Subjects:
Online Access:https://elifesciences.org/articles/52539
id doaj-e3efe8d8e0754d8bbbe2714fbaaca6ec
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Emma Hajaj
Galit Eisenberg
Shiri Klein
Shoshana Frankenburg
Sharon Merims
Inna Ben David
Thomas Eisenhaure
Sarah E Henrickson
Alexandra Chloé Villani
Nir Hacohen
Nathalie Abudi
Rinat Abramovich
Jonathan E Cohen
Tamar Peretz
Andre Veillette
Michal Lotem
spellingShingle Emma Hajaj
Galit Eisenberg
Shiri Klein
Shoshana Frankenburg
Sharon Merims
Inna Ben David
Thomas Eisenhaure
Sarah E Henrickson
Alexandra Chloé Villani
Nir Hacohen
Nathalie Abudi
Rinat Abramovich
Jonathan E Cohen
Tamar Peretz
Andre Veillette
Michal Lotem
SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
eLife
checkpoint
immunotherapy
cancer
T cells
author_facet Emma Hajaj
Galit Eisenberg
Shiri Klein
Shoshana Frankenburg
Sharon Merims
Inna Ben David
Thomas Eisenhaure
Sarah E Henrickson
Alexandra Chloé Villani
Nir Hacohen
Nathalie Abudi
Rinat Abramovich
Jonathan E Cohen
Tamar Peretz
Andre Veillette
Michal Lotem
author_sort Emma Hajaj
title SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
title_short SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
title_full SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
title_fullStr SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
title_full_unstemmed SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
title_sort slamf6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel t cell checkpoint
publisher eLife Sciences Publications Ltd
series eLife
issn 2050-084X
publishDate 2020-03-01
description SLAMF6 is a homotypic receptor of the Ig-superfamily whose exact role in immune modulation has remained elusive. Its constitutive expression on resting and activated T cells precludes it from being a bona fide exhaustion marker. By breeding Pmel-1 mice with SLAMF6 -/- mice, we generated donors for T cells lacking SLAMF6 and expressing a transgenic TCR for gp100-melanoma antigen. Activated Pmel-1xSLAMF6 -/- CD8+ T cells displayed improved polyfunctionality and strong tumor cytolysis. T-bet was the dominant transcription factor in Pmel-1 x SLAMF6 -/- cells, and upon activation, they acquired an effector-memory phenotype. Adoptive transfer of Pmel-1 x SLAMF6 -/- T cells to melanoma-bearing mice resulted in lasting tumor regression in contrast to temporary responses achieved with Pmel-1 T cells. LAG-3 expression was elevated in the SLAMF6 -/- cells, and the addition of the LAG-3-blocking antibody to the adoptive transfer protocol improved the SLAMF6 -/- T cells and expedited the antitumor response even further. The results from this study support the notion that SLAMF6 is an inhibitory immune receptor whose absence enables powerful CD8+ T cells to eradicate tumors.
topic checkpoint
immunotherapy
cancer
T cells
url https://elifesciences.org/articles/52539
work_keys_str_mv AT emmahajaj slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT galiteisenberg slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT shiriklein slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT shoshanafrankenburg slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT sharonmerims slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT innabendavid slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT thomaseisenhaure slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT sarahehenrickson slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT alexandrachloevillani slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT nirhacohen slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT nathalieabudi slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT rinatabramovich slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT jonathanecohen slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT tamarperetz slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT andreveillette slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
AT michallotem slamf6deficiencyaugmentstumorkillingandskewstowardaneffectorphenotyperevealingitasanoveltcellcheckpoint
_version_ 1721458553944801280
spelling doaj-e3efe8d8e0754d8bbbe2714fbaaca6ec2021-05-05T20:52:50ZengeLife Sciences Publications LtdeLife2050-084X2020-03-01910.7554/eLife.52539SLAMF6​ deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpointEmma Hajaj0https://orcid.org/0000-0003-2437-3146Galit Eisenberg1Shiri Klein2Shoshana Frankenburg3Sharon Merims4Inna Ben David5Thomas Eisenhaure6https://orcid.org/0000-0003-3999-3540Sarah E Henrickson7Alexandra Chloé Villani8Nir Hacohen9Nathalie Abudi10Rinat Abramovich11Jonathan E Cohen12Tamar Peretz13Andre Veillette14Michal Lotem15Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel; Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University, Jerusalem, IsraelSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, IsraelSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, IsraelSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, IsraelSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, IsraelSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, IsraelBroad Institute of MIT and Harvard, Cambridge, United StatesBroad Institute of MIT and Harvard, Cambridge, United States; Boston Children's Hospital, Department of Pediatrics, Boston, United StatesBroad Institute of MIT and Harvard, Cambridge, United States; Center for Cancer Research, Massachusetts General Hospital, Charlestown, United States; Department of Medicine, Harvard Medical School, Boston, United States; Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, United StatesBroad Institute of MIT and Harvard, Cambridge, United States; Center for Cancer Research, Massachusetts General Hospital, Charlestown, United States; Department of Medicine, Harvard Medical School, Boston, United StatesWohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel; Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, IsraelWohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel; Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, IsraelSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, IsraelSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, IsraelIRCM, Montreal Clinical Research Institute, Montreal, CanadaSharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel; Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University, Jerusalem, IsraelSLAMF6 is a homotypic receptor of the Ig-superfamily whose exact role in immune modulation has remained elusive. Its constitutive expression on resting and activated T cells precludes it from being a bona fide exhaustion marker. By breeding Pmel-1 mice with SLAMF6 -/- mice, we generated donors for T cells lacking SLAMF6 and expressing a transgenic TCR for gp100-melanoma antigen. Activated Pmel-1xSLAMF6 -/- CD8+ T cells displayed improved polyfunctionality and strong tumor cytolysis. T-bet was the dominant transcription factor in Pmel-1 x SLAMF6 -/- cells, and upon activation, they acquired an effector-memory phenotype. Adoptive transfer of Pmel-1 x SLAMF6 -/- T cells to melanoma-bearing mice resulted in lasting tumor regression in contrast to temporary responses achieved with Pmel-1 T cells. LAG-3 expression was elevated in the SLAMF6 -/- cells, and the addition of the LAG-3-blocking antibody to the adoptive transfer protocol improved the SLAMF6 -/- T cells and expedited the antitumor response even further. The results from this study support the notion that SLAMF6 is an inhibitory immune receptor whose absence enables powerful CD8+ T cells to eradicate tumors.https://elifesciences.org/articles/52539checkpointimmunotherapycancerT cells