RT Journal Article SR Electronic T1 Combined PD-L1 and TIM-3 blockade improves the expansion of fit human CD8+ antigen-specific T cells for adoptive immunotherapy JF medRxiv FD Cold Spring Harbor Laboratory Press SP 2021.08.30.21262835 DO 10.1101/2021.08.30.21262835 A1 Shirin Lak A1 Valérie Janelle A1 Anissa Djedid A1 Gabrielle Boudreau A1 Ann Brasey A1 Véronique Lisi A1 Cédric Carli A1 Lambert Busque A1 Vincent-Philippe Lavallée A1 Jean-Sébastien Delisle YR 2021 UL http://medrxiv.org/content/early/2021/10/10/2021.08.30.21262835.abstract AB Background The stimulation and expansion of antigen-specific T cells ex vivo enables the targeting of a multitude of cancer antigens. However, clinical scale T-cell expansion from rare precursors requires repeated stimulations ex vivo leading to T-cell terminal effector differentiation and exhaustion that adversely impact therapeutic potential. We leveraged immune checkpoint blockade relevant to antigen-specific CD8+ human T cells to improve the expansion and function of T cells targeting clinically relevant antigens.Methods A clinically-compliant protocol relying on peptide-pulsed monocyte-derived dendritic cells and cytokines was used to expand antigen-specific CD8+ targeting the oncogenic Epstein-Barr virus (EBV) and the tumor associated antigen (TAA) Wilms Tumor 1 (WT1) protein. The effects of antibody-mediated blockade of immune checkpoints applied to the cultures (T-cell expansion, phenotypes and function) were assessed at various time points. Genomic studies including single cell RNA (scRNA) sequencing and T-cell receptor sequencing were performed on EBV-specific T cells to inform about the impact of immune checkpoint blockade on the clonal distribution and gene expression of the expanded T cells.Results Several immune checkpoints were expressed early by ex vivo expanded antigen-specific CD8+ T cells, including PD-1 and TIM-3 with co-expression matching evidence of T-cell dysfunction as the cultures progressed. The introduction of anti-PD-L1 (expressed by the dendritic cells) and anti-TIM-3 antibodies in combination (but not individually) to the culture led to markedly improved antigen-specific T-cell expansion based on cell counts, fluorescent multimer staining and functional tests. This was not associated with evidence of T-cell dysfunction when compared to T cells expanded without immune checkpoint blockade. Genomics studies largely confirmed these results, showing that double blockade does not impart specific transcriptional programs or patterns on TCR repertoires. However, our data indicate that combined blockade may nonetheless alter gene expression in a minority of clonotypes and have donor-specific impacts.Conclusions The manufacturing of antigen-specific CD8+ T cells can be improved in terms of yield and functionality using blockade of TIM-3 and the PD-L1/PD-1 axis in combination. Overcoming the deleterious effects of multiple antigenic stimulations through PD-L1/TIM-3 blockade is a readily applicable approach for several adoptive-immunotherapy strategies.Competing Interest StatementThe authors have declared no competing interest.Funding StatementThis work was funded by the Leukemia/Lymphoma Society of Canada (grants #622735 and #430053) to JSD.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:All donors provided written informed consent, and all the experiments were approved by the Hema-Quebec and Hopital Maisonneuve-Rosemont Research Ethics Committees.I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesThe data can be found in the Gene Expression Omnibus (GSE182537 and GSE181682).ACTAdoptive Cell TherapyAPCAntigen Presenting CellCCR7C-C chemokine receptor type 7CDCluster of differentiationCDRComplementarity Determining RegionCTLA-4Cytotoxic T-lymphocyte-associated protein 4CTLsCytotoxic T-lymphocytesCTVCell Trace VioletCTYCell Trace YellowDCDendritic CellDMSODimethylsulfoxideEBVEpstein-Barr virusELISPOTEnzyme-linked immune absorbent spotFBSFetal Bovine SerumFOXP3Forkhead Box P3Gal-9Galectin-9GM-CSFGranulocyte-Macrophage Colony-Stimulating FactorGrzBGranzymeBHLAHuman Leukocyte AntigenIFNInterferonILInterleukinKLRG1Killer cell Lectin-like Receptor G1LAG-3Lymphocyte-Activation Gene 3LRSCLeukocyte Reduction System ChambermABMonoclonal AntibodyMHCMajor Histocompatibility ComplexNGSNext-generation SequencingPBMCPeripheral Blood Mononuclear CellPBSPhosphate-buffered salinePD-1Programmed Cell Death-1PD-L1Programmed Death Ligand-1PGE2Prostaglandin E2PHAT-cell mitogen phytohemagglutininPMAPhorbol 12-myristate 13-acetateRTRoom Temperaturesc-RNASingle Cell RNATAATumor-Associated AntigenTcmCentral Memory T cellTCRT-cell ReceptorTeffEffector T cellTemEffector Memory T cellTIM-3T-cell Immunoglobulin and Mucin-domain Containing-3TMB3,3′,5,5′-TetramethylbenzidineTNFTumor Necrosis FactorTSATumor-Specific AntigenWT1Wilms’ tumor suppressor gene1