[{"projectId":"cf35915c-dd80-459b-b315-7354892c00c3","title":"Characterization of maternal and fetal immunity following in utero spina bifida repair and surgery-induced preterm birth","investigator":"Enninga, Elizabeth Ann","investigatorInstitution":"Mayo Clinic","publicationName":"","researchArea":"Other","prettyUrls":{"365":"martinez-2025-s"},"prettyUrlList":["martinez-2025-s"],"summary":"Fetal surgery (FS) for in utero spina bifida (SB) repair has demonstrated\nimproved survival and reduced morbidity for infants; however, preterm birth (PTB) is\nreported in 36-80% of cases following SB repair. This prospective cohort study enrolled pregnant participants across three medical centers between 2022-2023. The FS group underwent fetoscopic SB repair between 24-26 weeks of gestation. A control group of low-risk pregnancies without SB were also recruited and matched by parity, gestational age at surgery, and fetal sex. Maternal blood samples were collected at baseline (before FS) and two weeks post-surgery (POD14). T cell receptor sequencing was completed on these samples.","prettyUrl":"martinez-2025-s","following":false,"created":"05/16/2025","featured":false,"publishedDate":"05/20/2025","urlOrId":"martinez-2025-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1cec4c1b-17b8-4037-8314-bc7cf1ee7831","title":"ABab-A2 vs ABab-I Mice-TCRα repertoire","investigator":"Dhamodaran, Arunraj","investigatorInstitution":"T-knife GmbH/MDC Berlin, Present Address: Microbiome and Cancer Division, German Cancer Research Center in the Helmholtz Association (DKFZ), Heidelberg, Germany.","publicationName":"Nature Communications","researchArea":"Cancer Immunotherapy","prettyUrls":{"364":"dhamodaran-2025-nc1"},"prettyUrlList":["dhamodaran-2025-nc1"],"summary":"TBD","prettyUrl":"dhamodaran-2025-nc1","following":false,"created":"05/16/2025","featured":false,"publishedDate":"05/16/2025","urlOrId":"dhamodaran-2025-nc1","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"3b68b71a-f58b-43ad-948e-133ea2dfc711","title":"ABab-A2 vs ABab-I Mice-TCRß repertoire","investigator":"Dhamodaran, Arunraj","investigatorInstitution":"Max Delbrück Center / T-knife GmbH, Present Address: Microbiome and Cancer Division, German Cancer Research Center in the Helmholtz Association (DKFZ), Heidelberg, Germany","publicationName":"Nature Communications","researchArea":"Cancer Immunotherapy","prettyUrls":{"363":"dhamodaran-2025-nc2"},"prettyUrlList":["dhamodaran-2025-nc2"],"summary":"TBD","prettyUrl":"dhamodaran-2025-nc2","following":false,"created":"05/16/2025","featured":false,"publishedDate":"05/16/2025","urlOrId":"dhamodaran-2025-nc2","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d3ad12ac-70db-4af0-8adc-0168dbb3ca4a","title":"γδ T-cell phenotype is impacted by CMV reactivation of patients short-term post-HSCT","investigator":"Uhlin, Michael","investigatorInstitution":"Department of Medicine, Huddinge, Karolinska Institutet","publicationName":"TBD","researchArea":"Other","prettyUrls":{"361":"hahn-2025-s"},"prettyUrlList":["hahn-2025-s"],"summary":"TBD","prettyUrl":"hahn-2025-s","following":false,"created":"05/16/2025","featured":false,"publishedDate":"05/16/2025","urlOrId":"hahn-2025-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"60246ee1-e0b5-40cd-a731-8dc42cbc02af","title":"The Molecular Epidemiology of Colorectal Cancer Study","investigator":"Gruber, Stephen B.","investigatorInstitution":"Department of Medical Oncology and Therapeutics Research","publicationName":"BMC Genomics","researchArea":"Cancer","prettyUrls":{"360":"schmit-2025-bmcg"},"prettyUrlList":["schmit-2025-bmcg"],"summary":"The Molecular Epidemiology of Colorectal Cancer Study (MECC) is a population-based study of incident CRC cases and healthy controls recruited from northern Israel from 1998 through 2017. After QC, 2,750 CRC cases with T cell receptor clonality and abundance data were derived from immunoSEQ assays.","prettyUrl":"schmit-2025-bmcg","following":false,"created":"05/08/2025","featured":false,"publishedDate":"05/15/2025","urlOrId":"schmit-2025-bmcg","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"db85fdb3-5a23-45ca-ba3d-499b9e64770d","title":"Locus folding mechanisms determine modes of antigen receptor gene assembly","investigator":"Bassing, Craig H.","investigatorInstitution":"Immunology Graduate Group, Perelman School of Medicine","publicationName":"JEM","researchArea":"Basic Immunology","prettyUrls":{"359":"allyn-2025-jem"},"prettyUrlList":["allyn-2025-jem"],"summary":"The dynamic folding of genomes regulates numerous biological processes, including antigen receptor (AgR) gene assembly. We show that, unlike other AgR loci, homotypic chromatin interactions and bidirectional chromosome looping both contribute to structuring Tcrb for efficient long-range V(D)J recombination. Inactivation of the CTCF binding element (CBE) or promoter at the most 5'Vβ segment (Trbv1) impaired loop extrusion originating locally and extending to DβJβ CBEs at the opposite end of Tcrb. Promoter or CBE mutation nearly eliminated Trbv1 contacts and decreased RAG endonuclease-mediated Trbv1 recombination. Importantly, Trbv1 rearrangement can proceed independent of substrate orientation, ruling out scanning by DβJβ-bound RAG as the sole mechanism of Vβ recombination, distinguishing it from Igh. Our data indicate that CBE-dependent generation of loops cooperates with promoter-mediated activation of chromatin to juxtapose Vβ and DβJβ segments for recombination through diffusion-based synapsis. Thus, the mechanisms that fold a genomic region can influence molecular processes occurring in that space, which may include recombination, repair, and transcriptional programming.","prettyUrl":"allyn-2025-jem","following":false,"created":"05/09/2025","featured":false,"publishedDate":"05/09/2025","urlOrId":"allyn-2025-jem","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"bd87e42c-4ad1-41dc-b375-c981d3527894","title":"Improving anti-CTLA-4 therapies through peptide masking and Fc non-fucosylation: preclinical characterization of three novel antibodies","investigator":"Jhatakia , Amy","investigatorInstitution":"Bristol Myers Squibb","publicationName":"TBD","researchArea":"Cancer Immunotherapy","prettyUrls":{"358":"jhatakia-2025-ccr"},"prettyUrlList":["jhatakia-2025-ccr"],"summary":"TBD","prettyUrl":"jhatakia-2025-ccr","following":false,"created":"04/15/2025","featured":false,"publishedDate":"04/15/2025","urlOrId":"jhatakia-2025-ccr","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"e854ec7c-0b49-4d75-a4da-ada06977a456","title":"Sex Differences in B Cells from the Joints of Children with Oligoarticular Juvenile Idiopathic Arthritis","investigator":"Henderson, Lauren","investigatorInstitution":"Division of Immunology, Boston Children’s Hospital, Harvard Medical School","publicationName":"Arthritis and Rheumatology","researchArea":"Other","prettyUrls":{"357":"lam-2025-arj"},"prettyUrlList":["lam-2025-arj"],"summary":"TBD","prettyUrl":"lam-2025-arj","following":false,"created":"03/17/2025","featured":false,"publishedDate":"03/18/2025","urlOrId":"lam-2025-arj","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a69126b7-b916-42fb-b815-7bbd58e249b0","title":"Radical hemithorax radiotherapy induces an increase in circulating PD-1 + T lymphocytes and in the soluble levels of PD-L1 in Malignant Pleural Mesothelioma patients: a possible synergy with PD-1/PD-L1 targeting treatment?","investigator":"Muraro Elena","investigatorInstitution":"Centro di Riferimento Oncologico di Aviano (CRO), IRCCS - Immunopathology and Cancer Biomarkers Unit, Department of Translational Research","publicationName":"Frontiers in Immunology","researchArea":"Cancer Immunotherapy","prettyUrls":{"356":"revelant-2025-s"},"prettyUrlList":["revelant-2025-s"],"summary":"Malignant Pleural Mesothelioma (MPM) is an aggressive tumor associated with asbestos exposure, characterized by a poor prognosis, managed with surgery, chemotherapy and radiotherapy. Recently, immunotherapy gives a survival advantage compared to chemotherapy, but limited to the non-epithelioid histotype, the rarest type. Radical hemithorax radiotherapy (RHRT) improves the Overall Survival (OS) of MPM patients, irrespective of histotype, and is able to induce immunomodulatory effects. In this study we aime to investigate changes in circulating T lymphocytes phenotype and activity, in MPM patients undergoing RHRT, to evaluate a possible therapeutic space for immunotherapy in this setting.\n\nTo assess immunomodulatory effects of RHRT we evaluate peripheral blood samples of 35 MPM patients collected before treatment, at the end of RT, and 1 month later. We first notice that higher Lymphocyte-to-Monocyte Ratio (LMR) levels, before RT, are associated with an improved OS. The immune monitoring performed by ELISA assays reveals a significant increase in the serum levels of sPD‐L1 and IFN‐γ at the end of RHRT. Furthermore, the percentage of PD‐1+ cells, evaluated by flow cytometry, significantly raise after RHRT in T cells, both CD4+ and CD8+. Also the proportion of proliferative cells is significantly expanded after RHRT in all T cell subtypes. After treatment we observe a significant increase in the number of patients showing WT-1 specific CD4+ T cells, measured by intracellular staining. The TCR repertoire analysis, investigated by Next Generation Sequencing, reveals an increased number of expanded T-cell clones after RHRT, and an association between TCR clonality and the percentage of proliferating cytotoxic T lymphocytes. The comparison of TCR sequences obtained in our cohort with those described in a literature cohort of MPM patients, reveals common entries, specific for MPM-associated antigens including WT-1.\n\nIn this setting, pre-treatment levels of LMR index seem to have a positive prognostic role, and RHRT would appear to induce immunomodulating effects, potential biomarkers for immunotherapy eligibility: i.e. increased PD-1+ T lymphocytes, proliferating T cells, expanded T cell clones and augmented levels of sPD-L1. These data suggest the design of a prospective study evaluating a maintenance immunotherapy after RHRT in MPM, even in the epithelioid histotype.","prettyUrl":"revelant-2025-s","following":false,"created":"03/14/2025","featured":false,"publishedDate":"03/17/2025","urlOrId":"revelant-2025-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1a2afc0b-6b64-45d3-a370-7d84ab42ed68","title":"Lymphocytic infiltration in stage II microsatellite stable colorectal tumors: A retrospective prognosis biomarker analysis","investigator":"Moreno, Victor","investigatorInstitution":"Catalan Institute of Oncology","publicationName":"PLOS Medicine","researchArea":"Cancer","prettyUrls":{"355":"sanz-pamplona-2025-plos"},"prettyUrlList":["sanz-pamplona-2025-plos"],"summary":"Background: Identifying stage II patients with colorectal cancer (CRC) at higher risk of progression is a clinical priority in order to optimize the advantages of adjuvant chemotherapy while avoiding unnecessary toxicity. Recently, the intensity and the quality of the host immune response in the tumor microenvironment have been reported to have an important role in tumorigenesis and an inverse association with tumor progression. This association is well established in microsatellite instable CRC. In this work, we aim to assess the usefulness of measures of T-cell infiltration as prognostic biomarkers in 640 stage II, CRC tumors, 582 of them confirmed microsatellite stable.\nMethods and findings: We measured both the quantity and clonality index of T cells by means of T-cell receptor (TCR) immunosequencing in a discovery dataset (95 patients with colon cancer diagnosed at stage II and microsatellite stable, median age 67, 30% women) and replicated the results in 3 additional series of stage II patients from 2 countries. Series 1 and 2 were recruited in Barcelona, Spain and included 112 fresh frozen (FF, median age 69, 44% women) and 163 formalin-fixed paraffin-embedded (FFPE, median age 67, 39% women) samples, respectively. Series 3 included 270 FFPE samples from patients recruited in Haifa, Northern Israel, as part of a large case-control study of CRC (median age 73, 46% women). Median follow-up time was 81.1 months. Cox regression models were fitted to evaluate the prognostic value of T-cell abundance and Simpson clonality of TCR variants adjusting by sex, age, tumor location, and stage (IIA and IIB). In the discovery dataset, higher TCR abundance was associated with better prognosis (hazard ratio [HR] for ≥Q1 = 0.25, 95% CI 0.10-0.63, P = 0.003). A functional analysis of gene expression on these tumors revealed enrichment in pathways related to immune response. Higher values of clonality index (lower diversity) were not associated with worse disease-free survival, though the HR for ≥Q3 was 2.32 (95% CI 0.90-5.97, P = 0.08). These results were replicated in an independent FF dataset (TCR abundance: HR = 0.30, 95% CI 0.12-0.72, P = 0.007; clonality: HR = 3.32, 95% CI 1.38-7.94, P = 0.007). Also, the association with prognosis was tested in 2 independent FFPE datasets. The same association was observed with TCR abundance (HR = 0.41, 95% CI 0.18-0.93, P = 0.03 and HR = 0.56, 95% CI 0.31-1, P = 0.042, respectively, for each FFPE dataset). However, the clonality index was associated with prognosis only in the FFPE dataset from Israel (HR = 2.45, 95% CI 1.39-4.32, P = 0.002). Finally, a combined analysis combining all microsatellite stable (MSS) samples demonstrated a clear prognosis value both for TCR abundance (HR = 0.39, 95% CI 0.26-0.57, P = 1.3e-06) and the clonality index (HR = 2.13, 95% CI 1.44-3.15, P = 0.0002). These associations were also observed when variables were considered continuous in the models (HR per log2 of TCR abundance = 0.85, 95% CI 0.78-0.93, P = 0.0002; HR per log2 or clonality index = 1.16, 95% CI 1.03-1.31, P = 0.016).\nLimitations: This is a retrospective study, and samples had been preserved with different methods. Validation series lack complete information about microsatellite instability (MSI) status and pathology assessment. The Molecular Epidemiology of Colorectal Cancer (MECC) study had information about overall survival instead of progression-free survival.\nConclusion: Results from this study demonstrate that tumor lymphocytes, assessed by TCR repertoire quantification based on a sequencing method, are an independent prognostic factor in microsatellite stable stage II CRC.","prettyUrl":"sanz-pamplona-2025-plos","following":false,"created":"02/27/2025","featured":false,"publishedDate":"03/01/2025","urlOrId":"sanz-pamplona-2025-plos","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c6f2c0bf-296a-4028-b5e6-3ee36f41de67","title":"Defining Resistance Mechanisms in TIL Therapy for NSCLC","investigator":"Creelan, Benjamin C.","investigatorInstitution":"Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute","publicationName":"TBD","researchArea":"Cancer","prettyUrls":{"353":"creelan-2025-s"},"prettyUrlList":["creelan-2025-s"],"summary":"TBD","prettyUrl":"creelan-2025-s","following":false,"created":"01/08/2025","featured":false,"publishedDate":"01/09/2025","urlOrId":"creelan-2025-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"190d24ea-d0ec-4296-b5d4-4cdff9ec7d59","title":"Graft-versus-host disease prophylaxis shapes T cell biology and immune reconstitution after hematopoietic cell transplant","investigator":"Kean, Leslie","investigatorInstitution":"Department of Pediatrics, Harvard Medical School, Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute","publicationName":"TBD","researchArea":"Cellular Therapy","prettyUrls":{"352":"siegel-2024-nm"},"prettyUrlList":["siegel-2024-nm"],"summary":"Abstract: Successful hematopoietic cell transplant requires immunosuppression to prevent graft-versus-host disease (GVHD), a lethal, T-cell-mediated post-transplant complication. The phase 3 BMT CTN 1703 trial demonstrated superior GVHD-free/relapse-free survival for post-transplant cyclophosphamide (PT-Cy)-based GVHD prophylaxis versus tacrolimus/methotrexate (Tac/MTX), but did not improve overall survival. To compare T-cell biology between GVHD prophylaxis regimens, 324 patients were co-enrolled onto BMT CTN 1801 (NCT03959241). We quantified T-cell immune reconstitution using multi-modal analysis, including T-cell receptor (TCR) sequencing of 2,359 longitudinal samples (180,432,350 T-cells). Compared to Tac/MTX, PT-Cy was associated with an early, substantial reduction in TCR diversity that was sustained for 2 years. PT-Cy led to a T-cell reconstitution bottleneck, including reduced thymic output and virus-associated TCRs. Decreased D+14 TCR diversity predicted prevention of chronic GVHD, but also correlated with increased moderate-to-severe infections. This study reveals how distinct immunosuppression strategies have significant effects on the global immune repertoire, underpinning post-transplant clinical outcomes.","prettyUrl":"siegel-2024-nm","following":false,"created":"12/23/2024","featured":false,"publishedDate":"12/27/2024","urlOrId":"siegel-2024-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"8378f7d6-196a-46b6-89d2-5fcf21a3baa7","title":"Chimeric antigen receptor macrophages (CAR-M) sensitize HER2+ solid tumors to PD1 blockade in pre-clinical models","investigator":"Pierini, Stefano","investigatorInstitution":"Carisma Therapeutics","publicationName":"Nature Communications","researchArea":"Cancer Immunotherapy","prettyUrls":{"351":"peirini-2024-nc"},"prettyUrlList":["peirini-2024-nc"],"summary":"We previously developed human CAR macrophages (CAR-M) and demonstrated redirection of macrophage anti-tumor function leading to tumor control in immunodeficient xenograft models. Here, we develop clinically relevant fully immunocompetent syngeneic models to evaluate the potential for CAR-M to remodel the tumor microenvironment (TME), induce T cell anti-tumor immunity, and sensitize solid tumors to PD1/PDL1 checkpoint inhibition. In vivo, anti-HER2 CAR-M significantly reduce tumor burden, prolong survival, remodel the TME, increase intratumoral T cell and natural killer (NK) cell infiltration, and induce antigen spreading. CAR-M therapy protects against antigen-negative relapses in a T cell dependent fashion, confirming long-term anti-tumor immunity. In HER2+ solid tumors with limited sensitivity to anti-PD1 (aPD1) monotherapy, the combination of CAR-M and aPD1 significantly improves tumor growth control, survival, and remodeling of the TME in pre-clinical models. These results demonstrate synergy between CAR-M and T cell checkpoint blockade and provide a strategy to potentially enhance response to aPD1 therapy for patients with non-responsive tumors","prettyUrl":"peirini-2024-nc","following":false,"created":"12/13/2024","featured":false,"publishedDate":"12/13/2024","urlOrId":"peirini-2024-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"2439ae7e-5c9c-4149-8c9f-8ff65ec0db7d","title":"Pre-Transplant T-Cell Clonal Analysis Identifies CD8+ Donor Reactive Clones That Contribute To Kidney Transplant Rejection Following Two Different Induction Modalities","investigator":"Mathew, James","investigatorInstitution":"Northwestern Universty Feinberg School of Medicine","publicationName":"Frontiers in Immunology","researchArea":"Organ transplant","prettyUrls":{"350":"sanders-2024-stm"},"prettyUrlList":["sanders-2024-stm"],"summary":"Introduction: Responses to allogeneic human leukocyte antigen (HLA) molecules limit the survival of transplanted organs. The changes in T-cell alloreactivity that contribute to this process, however, are not fully understood. We defined a set of donor reactive T-cell clones (DRTC) with the goal to elucidate signatures of kidney allograft rejection.\n\nMethods: DRTC were identified pretransplant using an anti-donor mixed lymphocyte reaction assay: CFSE-diluting CD4+ and CD8+ DRTC were flow-sorted, and the TCR sequences were identified using Adaptive Immunosequencing. DRTC were then tracked in post-transplant biopsies, blood, and urine samples in a cohort of kidney transplant recipients. \n\nResults: In patients with an abnormal biopsy, the majority of CD8+ DRTC found within the allograft were detected in the circulating pre-transplant repertoire. Circulating CD8+ DRTC were more abundant pre- and post-transplant in patients that received non-lymphodepletional induction and developed an abnormal biopsy when compared to stable patients. Additionally, DRTC were detected as early as two weeks post-transplant in the urine of some patients, with some of these clones subsequently identified in follow-up kidney biopsy samples.\n\nDiscussion: The findings of our study add to our understanding of T-cell alloreactivity following kidney transplantation and provide evidence for the role of pre-defined alloreactive T-cells in the development of allograft rejection.","prettyUrl":"sanders-2024-stm","following":false,"created":"08/13/2024","featured":false,"publishedDate":"12/06/2024","urlOrId":"sanders-2024-stm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"0f19e35e-fedd-4157-9ea4-cccd7cfefc7d","title":"Intratumoral T-cell receptor repertoire composition predicts overall survival in patients with pancreatic ductal adenocarcinoma","investigator":"Hogg, Graham","investigatorInstitution":"Washington University School of Medicine in St. Louis","publicationName":"Oncoimmunology","researchArea":"Cancer","prettyUrls":{"349":"pothuri-2024-oi"},"prettyUrlList":["pothuri-2024-oi"],"summary":"Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy that is refractory to immune checkpoint inhibitor therapy. However, intratumoral T-cell infiltration correlates with improved overall survival (OS). Herein, we characterized the diversity and antigen specificity of the PDAC T-cell receptor (TCR) repertoire to identify novel immune-relevant biomarkers. Demographic, clinical, and TCR-beta sequencing data were collated from 353 patients across three cohorts that underwent surgical resection for PDAC. TCR diversity was calculated using Shannon Wiener index, Inverse Simpson index, and “True entropy.” Patients were clustered by shared repertoire specificity. TCRs predictive of OS were identified and their associated transcriptional states were characterized by single-cell RNAseq. In multivariate Cox regression models controlling for relevant covariates, high intratumoral TCR diversity predicted OS across multiple cohorts. Conversely, in peripheral blood, high abundance of T-cells, but not high diversity, predicted OS. Clustering patients based on TCR specificity revealed a subset of TCRs that predicts OS. Interestingly, these TCR sequences were more likely to encode CD8+ effector memory and CD4+ T-regulatory (Tregs) T-cells, all with the capacity to recognize beta islet-derived autoantigens. As opposed to T-cell abundance, intratumoral TCR diversity was predictive of OS in multiple PDAC cohorts, and a subset of TCRs enriched in high-diversity patients independently correlated with OS. These findings emphasize the importance of evaluating peripheral and intratumoral TCR repertoires as distinct and relevant biomarkers in PDAC.","prettyUrl":"pothuri-2024-oi","following":false,"created":"11/13/2024","featured":false,"publishedDate":"11/19/2024","urlOrId":"pothuri-2024-oi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"6f9fb8df-7215-47fc-9d54-7f52d973b8ee","title":"TBD","investigator":"Elyanow, Rebecca","investigatorInstitution":"Adaptive Biotechnologies","publicationName":"TBD","researchArea":"Autoimmune Disorders","prettyUrls":{"348":"elyanow-2024-s"},"prettyUrlList":["elyanow-2024-s"],"summary":"TBD","prettyUrl":"elyanow-2024-s","following":false,"created":"11/05/2024","featured":false,"publishedDate":"11/07/2024","urlOrId":"elyanow-2024-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d300d36c-2166-4525-b45b-95085e1149f1","title":"Clonal analysis of SepSecS-specific B and T cells in autoimmune hepatitis","investigator":"Sallusto, Federica","investigatorInstitution":"Institute for Research in Biomedicine (IRB)","publicationName":"Journal of Clinical Investigation","researchArea":"Autoimmune Disorders","prettyUrls":{"347":"kramer-2024-jci"},"prettyUrlList":["kramer-2024-jci"],"summary":"TBD","prettyUrl":"kramer-2024-jci","following":false,"created":"10/29/2024","featured":false,"publishedDate":"10/29/2024","urlOrId":"kramer-2024-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"4f73c166-de99-439a-9594-1bd1b0e699ce","title":"Tumor-resident Lactobacillus iners confers chemoradiation resistance through lactate-induced metabolic rewiring","investigator":"Colbert, Lauren E.","investigatorInstitution":"The University of Texas M.D. Anderson Cancer Center","publicationName":"Cancer Cell","researchArea":"Cancer","prettyUrls":{"346":"colbert-2024-cc"},"prettyUrlList":["colbert-2024-cc"],"summary":"Tumor microbiota can produce active metabolites that affect cancer and immune cell signaling, metabolism, and proliferation. Here, we explore tumor and gut microbiome features that affect chemoradiation response in cervical cancer patients through a combined approach of deep microbiome sequencing, sequential, targeted bacterial culture and in vitro assays. We identify that an obligate L-lactate producing lactic acid bacteria found in tumors, Lactobacillus iners, is associated with decreased survival in patients, induces chemotherapy and radiation resistance in cervical cancer cells, and leads to metabolic rewiring of tumors. Genomically similar L-lactate producing lactic acid bacteria commensal to other body sites are also significantly associated with survival in colorectal cancer, lung cancer, head and neck cancer and skin cancer. Our findings demonstrate that lactic acid bacteria in the tumor microenvironment can alter tumor metabolism and lactate signaling pathways to cause therapy resistance. Lactic acid bacteria could be promising therapeutic targets across cancer types.","prettyUrl":"colbert-2024-cc","following":false,"created":"10/22/2024","featured":false,"publishedDate":"10/22/2024","urlOrId":"colbert-2024-cc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"63145368-9b39-4cd1-b9bf-88e3eb7396cf","title":"A phase I clinical trial of intrahepatic artery delivery of TG6002 in combination with oral 5-fluorocytosine in patients with liver-dominant metastatic colorectal cancer","investigator":"West, Emma","investigatorInstitution":"Leeds Institute of Medical Research at St. James’s, Translational Cancer Immunotherapy Group","publicationName":"TBD","researchArea":"Cancer Immunotherapy","prettyUrls":{"345":"west-2024-s"},"prettyUrlList":["west-2024-s"],"summary":"Background: Effective treatment for patients with metastatic cancer is limited, particularly for colorectal cancer patients with metastatic liver lesions (mCRC), where accessibility to numerous tumours is essential for favorable clinical outcomes. Oncolytic viruses (OVs) selectively replicate in cancer cells; however, direct targeting of inaccessible lesions is limited when using conventional intravenous or intratumoural administration routes. \nMethods: We conducted a multi-centre, dose-escalation, phase I study of vaccinia virus, TG6002, via intrahepatic artery (IHA) delivery in combination with the oral pro-drug 5-fluorocytosine to fifteen mCRC patients. \nResults: Successful IHA delivery of replication-competent TG6002 was achieved, as demonstrated by virus within tumour biopsies. Functional transcription of the FCU1 transgene indicates viral replication within the tumour, with higher plasma 5-fluorouracil associated with patients receiving the highest dose of TG6002. IHA delivery of TG6002 correlated with a robust systemic peripheral immune response to virus with activation of peripheral blood mononuclear cells, associated with a proinflammatory cytokine response and release of calreticulin, potentially indicating immunogenic cell death. Gene Ontology analyses of differentially-expressed genes reveal a significant immune response at the transcriptional level in response to treatment. Moreover, an increase in the number and frequency of T-cell receptor clones against both cancer- and neo-antigens, with elevated functional activity, may be associated with improved anti-cancer activity. Despite these findings, no clinical efficacy was observed. \nConclusions: In summary, these data demonstrate delivery of OV to tumour via IHA administration, associated with viral replication and significant peripheral immune activation. Collectively, the data supports the need for future studies using IHA administration of OVs.","prettyUrl":"west-2024-s","following":false,"created":"10/16/2024","featured":false,"publishedDate":"10/16/2024","urlOrId":"west-2024-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"30f6e6c6-cdee-4d15-abeb-81ff75813086","title":"IL-12 drives the expression of the inhibitory receptor NKG2A on human tumor-reactive CD8+ T cells","investigator":"Duhen, Thomas","investigatorInstitution":"Earle A. Chiles Research Institute, Providence Cancer Institute","publicationName":"TBD","researchArea":"Cancer Immunotherapy","prettyUrls":{"344":"fesneau-2024-nc"},"prettyUrlList":["fesneau-2024-nc"],"summary":"Blockade of NKG2A/HLA-E interaction is a promising strategy to unleash the anti-tumor response. Yet the role of NKG2A+ CD8+ T cells in the anti-tumor response and the regulation of NKG2A expression are still poorly understood. Here, by performing CITE-seq on human T cells derived from head and neck squamous cell carcinoma and colorectal cancer, we show that NKG2A expression is induced in tumor-infiltrating CD8+ T cells differentiating into cytotoxic, CD39+CD103+ double positive (DP) T cells. This developmental trajectory lead to TCR repertoire overlap between the NKG2A– and NKG2A+ DP CD8 T cells, suggesting shared antigen specificities. Mechanistically, IL-12 is essential for the expression of the NKG2A on naïve CD8+ T cells in a CD40/CD40L- dependent manner, in conjunction with TCR stimulation and increased TGF-β levels. Our study thus reveals that NKG2A is induced by IL-12 on human tumor-reactive CD8+ T cells exposed to a TGF-b-rich environment, highlighting an underappreciated immuno-regulatory feedback loop dependent on IL-12 stimulation.","prettyUrl":"fesneau-2024-nc","following":false,"created":"10/07/2024","featured":false,"publishedDate":"10/07/2024","urlOrId":"fesneau-2024-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"8fe07a3e-a1a4-4b06-8481-b909a1b9c1c8","title":"Ongoing replication stress tolerance and clonal T cell responses distinguish liver and lung recurrence and outcomes in pancreatic cancer","investigator":"Sears, Rosalie C.","investigatorInstitution":"Department of Molecular and Medical Genetics, Oregon Health and Science University","publicationName":"Nature Cancer","researchArea":"Cancer","prettyUrls":{"343":"link-2023-nc"},"prettyUrlList":["link-2023-nc"],"summary":"Metastatic pancreatic ductal adenocarcinoma (mPDAC) is lethal, yet a subset of patients who have metastatic disease that spreads to the lung, but not the liver, survive longer. We generated large genomic, transcriptomic and T cell receptor sequencing datasets, along with deep clinical annotation, to elucidate unique tumor and immune features that distinguish patients who develop metastases in the liver (liver cohort) versus those with lung-avid but liver-averse mPDAC (lung cohort). Lung cohort patients had better survival outcomes than liver cohort patients even within the same tumor subtype. Using a novel gene signature for liver versus lung organotropism in primary tumors independent from known tumor subtypes (pORG), we identified ongoing replication stress (RS) response pathways in high pORG/liver cohort tumors and greater density of leukocytes in low pORG/lung cohort tumors. Patients with low pORG tumors survived longer, especially if their tumors had alterations in DNA damage repair genes. Patients that are in the lung cohort demonstrated new T cell clonal development in their primary and metastatic tumors leading to diverse peripheral blood TCR repertoires. Our study demonstrates that liver-avid metastatic PDAC is associated with an ongoing RS response, whereas tumors lacking the RS response with ongoing T cell clonal responses may have unique vulnerabilities allowing long-term survival in patients with lung-avid, liver-averse metastatic PDAC.","prettyUrl":"link-2023-nc","following":false,"created":"02/08/2023","featured":false,"publishedDate":"09/24/2024","urlOrId":"link-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7d481cd7-1cf4-4612-8cbd-b33b333ccfd9","title":"Neoadjuvant or concurrent Anti PD-L1 (Atezolizumab) with chemoradiation for locally advanced cervical cancer","investigator":"Zamarin, Dmitriy","investigatorInstitution":"Icahn School of Medicine at Mount Sinai","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"340":"mayadev-2024-nc"},"prettyUrlList":["mayadev-2024-nc"],"summary":"Combination of immune checkpoint blockade (ICB) with chemoradiation (CRT) is now approved in patients with locally advanced cervical cancer (LACC) but optimal sequencing of CRT and ICB is unknown. NRG-GY017 (NCT03738228) was a randomized phase I trial of differential sequencing of atezolizumab and CRT in patients with high-risk node-positive LACC. The primary objective was to evaluate the expansion of tumor-associated T-cell receptor (TCR) clones in peripheral blood as a surrogate measure of anti-tumor immune response. Secondary objectives included toxicity and 2-year disease-free survival (DFS). Forty patients were randomized and 36 received 3 doses of atezolizumab neoadjuvant and concurrent with CRT (Arm A) vs. concurrent with CRT (Arm B). After cycle 1, there was peripheral expansion of higher proportion of tumor-associated TCR clones in Arm A than in Arm B (median 0.39 vs. 0.06, p=0.0025) that remained numerically higher at the day 21 primary endpoint (p=0.052 by 2-sample t test). At the median follow up of 25.8 months, 2-year DFS was 76% in Arm A and 56% in Arm B (p=0.28). There were no new safety signals. In conclusion, addition of neoadjuvant ICB prior to CRT was safe and was associated with immunologically and clinically favorable outcomes, warranting larger confirmatory studies.","prettyUrl":"mayadev-2024-nc","following":false,"created":"09/16/2024","featured":false,"publishedDate":"09/16/2024","urlOrId":"mayadev-2024-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"8ddebffc-242e-48e2-a84c-9c8ec6584e89","title":"Late-stage tertiary lymphoid structures in hepatocellular carcinoma treated with neoadjuvant immune checkpoint blockade","investigator":"Shu, Daniel","investigatorInstitution":"University of Maryland","publicationName":"TBD","researchArea":"Cancer Immunotherapy","prettyUrls":{"339":"shu-2024-ni"},"prettyUrlList":["shu-2024-ni"],"summary":"Coming soon","prettyUrl":"shu-2024-ni","following":false,"created":"09/09/2024","featured":false,"publishedDate":"09/10/2024","urlOrId":"shu-2024-ni","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1b8516df-d9ad-49f4-ac56-a661e6456830","title":"AVATAR mice for personalized medicine in patients with atopic dermatitis","investigator":"Kim, Hye Li","investigatorInstitution":"Yonsei university","publicationName":"TBD","researchArea":"Other","prettyUrls":{"338":"park-2024-s"},"prettyUrlList":["park-2024-s"],"summary":"TBD","prettyUrl":"park-2024-s","following":false,"created":"09/06/2024","featured":false,"publishedDate":"09/06/2024","urlOrId":"park-2024-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"945aadf7-e2ad-4a79-93b7-f013f9d9ba1b","title":"Generation of antigen-specific memory CD4 T cells by heterologous immunization enhances the magnitude of the germinal center response upon influenza infection","investigator":"Sircy, Linda","investigatorInstitution":"University of Utah","publicationName":"TBD","researchArea":"Other","prettyUrls":{"337":"williams-2024-p"},"prettyUrlList":["williams-2024-p"],"summary":"Current influenza vaccine strategies have yet to overcome significant obstacles, including rapid antigenic drift of seasonal influenza viruses, in generating efficacious long-term humoral immunity. Due to the necessity of germinal center formation in generating long-lived high affinity antibodies, the germinal center has increasingly become a target for the development of novel or improvement of less-efficacious vaccines. However, there remains a major gap in current influenza research to effectively target T follicular helper cells during vaccination to alter the germinal center reaction. In this study, we used a heterologous infection or immunization priming strategy to seed an antigen-specific memory CD4+ T cell pool prior to influenza infection in mice to evaluate the effect of recalled memory T follicular helper cells in increased help to influenza-specific primary B cells and enhanced generation of neutralizing antibodies. We found that heterologous priming with intranasal infection with acute lymphocytic choriomeningitis virus (LCMV) or intramuscular immunization with adjuvanted recombinant LCMV glycoprotein induced increased antigen-specific effector CD4+ T and B cellular responses following infection with a recombinant influenza strain that expresses LCMV glycoprotein. Heterologously primed mice had increased expansion of secondary Th1 and Tfh cell subsets, including increased CD4+ TRM cells in the lung. However, the early enhancement of the germinal center cellular response following influenza infection did not impact influenza-specific antibody generation or B cell repertoires compared to primary influenza infection. Overall, our study suggests that while heterologous infection or immunization priming of CD4+ T cells is able to enhance the early germinal center reaction, further studies to understand how to target the germinal center and CD4+ T cells specifically to increase long-lived antiviral humoral immunity are needed.","prettyUrl":"williams-2024-p","following":false,"created":"08/27/2024","featured":false,"publishedDate":"08/28/2024","urlOrId":"williams-2024-p","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"362fe554-59cd-469e-81d1-c87768ef434d","title":"Phase 1 study combining elotuzumab with autologous stem cell transplant and lenalidomide for multiple myeloma","investigator":"Kim-schulze, Seunghee","investigatorInstitution":"The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mt. Sinai","publicationName":"Journal for ImmunoTherapy of Cancer","researchArea":"Cancer","prettyUrls":{"335":"coffey-2024-jitc"},"prettyUrlList":["coffey-2024-jitc"],"summary":"Background: Autologous stem cell transplantation (ASCT) after induction therapy improves disease-free survival for patients with multiple myeloma (MM). While the goal of ASCT is to render a minimal disease state, it is also associated with eradication of immunosuppressive cells, and we hypothesize that early introduction of immunotherapy post-ASCT may provide a window of opportunity to boost treatment efficacy.\n\nMethods: We conducted a phase 1 clinical trial to investigate the application of autologous lymphocyte infusion and anti-SLAMF7 monoclonal antibody, elotuzumab, after ASCT in patients with newly diagnosed MM previously treated with induction therapy. In addition to CD34+ stem cells, peripheral blood mononuclear cells were harvested prior to transplant and infused on day 3 after stem cell infusion to accelerate immune reconstitution and provide autologous natural killer (NK) cells that are essential to the mechanism of elotuzumab. Elotuzumab was administered starting on day 4 and then every 28 days after until 1 year post-ASCT. Cycles 4–12 were administered with standard-of-care lenalidomide maintenance.\n\nResults: All subjects were evaluated for safety, and 13 of 15 subjects completed the treatment protocol. At 1 year post-ASCT, the disease status of enrolled subjects was as follows: five stringent complete responses, one complete response, six very good partial responses, one partial response, and two progressive diseases. The treatment plan was well tolerated, with most grade 3 and 4 AEs being expected hematologic toxicities associated with ASCT. Correlative analysis of the immune microenvironment demonstrated a trend toward reduced regulatory T cells during the first 3 months post-transplant followed by an increase in NK cells and monocytes in patients achieving a complete remission.\n\nConclusions: This phase 1 clinical trial demonstrates that early introduction of immunotherapy after ASCT is well tolerated and shows promising disease control in patients with MM, accompanied by favorable changes in the immune microenvironment.","prettyUrl":"coffey-2024-jitc","following":false,"created":"08/12/2024","featured":false,"publishedDate":"08/12/2024","urlOrId":"coffey-2024-jitc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"737666e5-ffcf-4e0e-b51a-30ff5ad34405","title":"Xenogeneic Graft-Versus-Host Disease in Humanized NSG and NSG-HLA-A2/HHD Mice","investigator":"Ehx, Gregory","investigatorInstitution":"GIGA Institute: Hematology","publicationName":"Frontiers in Immunology","researchArea":"Adoptive T Cell Therapy","prettyUrls":{"334":"ehx-2024-ji"},"prettyUrlList":["ehx-2024-ji"],"summary":"Despite the increasing use of humanized mouse models to study new approaches of graft-versus-host disease (GVHD) prevention, the pathogenesis of xenogeneic GVHD (xGVHD) in these models remains misunderstood. The aim of this study is to describe this pathogenesis in NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice infused with human PBMCs and to assess the impact of the expression of HLA-A0201 by NSG mice cells (NSG-HLA-A2/HHD mice) on xGVHD and graft-versus-leukemia (GvL) effects, by taking advantage of next-generation technologies. We found that T cells recovered from NSG mice after transplantation had upregulated expression of genes involved in cell proliferation, as well as in TCR, co-stimulatory, IL-2/STAT5, mTOR and Aurora kinase A pathways. T cells had mainly an effector memory or an effector phenotype and exhibited a Th1/Tc1-skewed differentiation. TCRβ repertoire diversity was markedly lower both in the spleen and lungs (a xGVHD target organ) than at infusion. There was no correlation between the frequencies of specific clonotypes at baseline and in transplanted mice. Finally, expression of HLA-A0201 by NSG mice led to more severe xGVHD and enhanced GvL effects toward HLA-A2+ leukemic cells. Altogether our data demonstrate that the pathogenesis of xGVHD shares important features with human GVHD and that NSG-HLA-A2/HHD mice could serve as better model to study GVHD and GvL effects.","prettyUrl":"ehx-2024-ji","following":false,"created":"07/23/2024","featured":false,"publishedDate":"07/24/2024","urlOrId":"ehx-2024-ji","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"eaa3e269-ee08-4a6b-965a-50f228cdc2a4","title":"IL-7-dependent and -independent lineages of IL-7R-dependent human T cells","investigator":"Puel, A","investigatorInstitution":"Institut IMAGINE","publicationName":"Journal of Clinical Investigation","researchArea":"Basic Immunology","prettyUrls":{"333":"arango-franco-2024-jci"},"prettyUrlList":["arango-franco-2024-jci"],"summary":"TBD","prettyUrl":"arango-franco-2024-jci","following":false,"created":"06/19/2024","featured":false,"publishedDate":"06/21/2024","urlOrId":"arango-franco-2024-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"394ed703-0279-4982-8213-e2dbbc12c136","title":"COVID-19 vaccinated Children, Adolescents, and Young Adults with Acute Lymphoblastic Leukemia show Spike reactive antibodies and multifunctional T-cells","investigator":"Parekh, Chintan","investigatorInstitution":"Children's Hospital Los Angeles","publicationName":"TBD","researchArea":"Vaccine efficacy","prettyUrls":{"332":"parker-2024-s"},"prettyUrlList":["parker-2024-s"],"summary":"Little is known about the efficacy of COVID-19 vaccines during acute lymphoblastic leukemia therapy (ALL); data for COVID-19 vaccine immune responses in pediatric leukemia remain sparse. We used flow cytometry and TCR sequencing to assess Spike Reactive T-cell responses in 20 patients aged 5-25 years undergoing Acute lymphoblastic leukemia (ALL) chemotherapy who had received COVID-19 vaccination. TCR sequencing was also done on T-cells from 5 COVID-19 vaccinated healthy adult individuals (healthy controls). Spike reactive T-cells (SRT) were detected by flow cytometry in 9 of 20 (45%) vaccinated patients undergoing ALL chemotherapy. Sequencing revealed public CD4+ and CD8+ TCR sequences reactive to epitopes across the Spike Protein in these patients. In conclusion, COVID-19 vaccination induced T-cell responses in a substantial fraction of children and young adults undergoing ALL chemotherapy.","prettyUrl":"parker-2024-s","following":false,"created":"06/07/2024","featured":false,"publishedDate":"06/10/2024","urlOrId":"parker-2024-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"fc8472cd-aebc-4b40-b8e7-37f47a9b170f","title":"Expansion of the tissue-based T-cell receptor repertoire is distinct from the PBMC response after immunotherapeutic HSV-2 vaccine","investigator":"Ford, Emily S","investigatorInstitution":"Vaccine and Infectious Diseases Division and Division of Allergy and Infectious Diseases","publicationName":"TBD","researchArea":"Response to Therapeutic Agent","prettyUrls":{"331":"ford-2024-jci"},"prettyUrlList":["ford-2024-jci"],"summary":"Coming Soon...","prettyUrl":"ford-2024-jci","following":false,"created":"06/06/2024","featured":false,"publishedDate":"06/10/2024","urlOrId":"ford-2024-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"98c4a4eb-fa62-48d5-8637-170f0a86b0ab","title":"Differential tumor immune microenvironment coupled with tumor progression or tumor eradication in HPV-antigen expressing squamous cell carcinoma (SCC) models.","investigator":"Wang, Jing","investigatorInstitution":"Department of Medicine, University of Pittsburgh","publicationName":"Frontiers in Immunology","researchArea":"Cancer","prettyUrls":{"330":"shivarudrappa-2024-fi"},"prettyUrlList":["shivarudrappa-2024-fi"],"summary":"Human papilloma virus (HPV) is an etiological factor of head and neck squamous cell carcinoma (HNSCC). To investigate the role of HPV antigen in anti-tumor immunity, we established mouse models by expressing HPV16 E6 and E7 in a SCC tumor cell line. We obtained two HPV antigen-expressing clones (C-225 and C-100) transplantable into C57BL/6 recipients. We found that C-225 elicited complete eradication in C57BL/6 mice (eradicated), whereas C-100 grew progressively (growing). We examined immune tumor microenvironment (TME) using flow cytometry and found that eradicated or growing tumors exhibited differential immune profiles that may influence the outcome of anti-tumor immunity. Surprisingly, the percentage of CD8 and CD4 tumor-infiltrating lymphocytes (TILs) was much higher in growing (C-100) than eradicated (C-225) tumor. However, the TILs upregulated PD-1 and LAG-3 more potently and exhibited impaired effector functions in growing tumor compared to their counterparts in eradicated tumor. C-225 TME is highly enriched with myeloid cells, especially polymorphonuclear (PMN) myeloid-derived suppressor cells (MDSC), whereas the percentage of M-MDSC and tumor-associated macrophages (TAMs) was much higher in C-100 TME, especially M2-TAMs (CD206+). The complete eradication of C-225 depended on CD8 T cells and elicited anti-tumor memory responses upon secondary tumor challenge. We employed DNA sequencing to identify differences in the T cell receptor of peripheral blood lymphocytes pre- and post-secondary tumor challenge. Lastly, C-225 and C-100 tumor lines harbored different somatic mutations. Overall, we uncovered differential immune TME that may underlie the divergent outcomes of anti-tumor immunity by establishing two SCC tumor lines, both of which express HPV16 E6 and E7 antigens. Our experimental models may provide a platform for pinpointing tumor-intrinsic versus host-intrinsic differences in orchestrating an immunosuppressive TME in HNSCCs and for identifying new targets that render tumor cells vulnerable to immune attack.","prettyUrl":"shivarudrappa-2024-fi","following":false,"created":"06/04/2024","featured":false,"publishedDate":"06/04/2024","urlOrId":"shivarudrappa-2024-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c5a35918-da0a-49fd-8ec5-8c022554aeca","title":"IgH and IgL data for MRD tracking post-CD20 CAR T cell therapy in a patient with mantle cell lymphoma","investigator":"Till, Brian","investigatorInstitution":"Fred Hutchinson Cancer Center","publicationName":"Blood Cancer Discovery","researchArea":"Adoptive T Cell Therapy","prettyUrls":{"329":"mo-2024-3-bcd"},"prettyUrlList":["mo-2024-3-bcd"],"summary":"A 62 year old man with mantle cell lymphoma was treated with cyclophosphamide (CY) lymphodepletion, 3 escalating doses of CD20-targeted CAR T cell infusions, and IL-2 injections as per Till et al, Blood 2012; 119(17):3940-50. He was re-treated with CY, CAR-T (1 dose only) and IL-2 after relapse 2 years later, had transient apparent progression at 6 weeks post CAR-T infusion, followed by a remission a few weeks later, and remained in remission for several years. These samples provide data for MRD tracking. The samples are as follows:\n\n02583-01OH-207791: Tumor biopsy from 48h post his 3rd CAR-T infusion (IGH)\n02583-01OL-207791: Tumor biopsy from 48h post his 3rd CAR-T infusion (IGL)\n02583-02BH: Baseline #2 (before re-treatment with 4th CAR T cell infusion) (IGH)\n02583-02BL: Baseline #2 (before re-treatment with 4th CAR T cell infusion) (IGH)\n02583-03BH-1: 1.7 months post the 4th CAR-T infusion (IGH)\n02583-03BL-1: 1.7 months post the 4th CAR-T infusion (IGL)\n02583-03BH: 6.4 months post the 4th CAR-T infusion (IGH)\n02583-03BL: 6.4 months post the 4th CAR-T infusion (IGL)\n02583-05BH: 64 months post the 4th CAR-T infusion (IGH)\n02583-05BL: 64 months post the 4th CAR-T infusion (IGL)","prettyUrl":"mo-2024-3-bcd","following":false,"created":"05/03/2024","featured":false,"publishedDate":"05/07/2024","urlOrId":"mo-2024-3-bcd","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"859cc661-77b7-4900-9671-c7d673c8ba30","title":"B cell clonality data over time post-CD20 CAR-T in a follicular lymphoma patient with late remission","investigator":"Till, Brian","investigatorInstitution":"Fred Hutchinson Cancer Center","publicationName":"Blood Cancer Discovery","researchArea":"Adoptive T Cell Therapy","prettyUrls":{"328":"mo-2024-2-bcd"},"prettyUrlList":["mo-2024-2-bcd"],"summary":"A 28 year-old patient was treated with cyclophosphamide lymphodepletion, 3 escalating doses of CD20-targeted CAR T cells (as per Till et al, Blood 2012; 119(17):3940-50), with a delayed partial response around 3 months and then a complete response around 2.5 years, suggesting an endogenous anti-tumor response. This project contains the IgH and IgK data from his tumor biopsy and at baseline and at 3 years, demonstrating resolution of two prominent subclones but residual levels of one subclone (MRD) at 3 years post-treatment.","prettyUrl":"mo-2024-2-bcd","following":false,"created":"05/03/2024","featured":false,"publishedDate":"05/07/2024","urlOrId":"mo-2024-2-bcd","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"05fbfd79-e689-43b3-97cc-2d63547ebf4d","title":"TCR-beta repertoire over time post-CD20 CAR-T in a follicular lymphoma patient with late remission","investigator":"Till, Brian","investigatorInstitution":"Fred Hutchinson Cancer Center","publicationName":"Blood Cancer Discovery","researchArea":"Adoptive T Cell Therapy","prettyUrls":{"327":"mo-2024-1-bcd"},"prettyUrlList":["mo-2024-1-bcd"],"summary":"A 28 year-old patient was treated with cyclophosphamide lymphodepletion, 3 escalating doses of CD20-targeted CAR T cells (as per Till et al, Blood 2012; 119(17):3940-50), with a delayed partial response around 3 months and then a complete response around 2.5 years, suggesting an endogenous anti-tumor response.","prettyUrl":"mo-2024-1-bcd","following":false,"created":"05/03/2024","featured":false,"publishedDate":"05/07/2024","urlOrId":"mo-2024-1-bcd","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"0518c8a6-b11d-4865-97f9-49906f557723","title":"CLN-617 retains IL-2 and IL-12 in injected tumors to drive robust and systemic immune-mediated anti-tumor activity","investigator":"Rakhra, Kavya","investigatorInstitution":"Cullinan Oncology","publicationName":"Cancer Immunology Research","researchArea":"Cancer Immunotherapy","prettyUrls":{"326":"mehta-2024-cir"},"prettyUrlList":["mehta-2024-cir"],"summary":"Despite clinical evidence of anti-tumor activity, cytokine therapies have been hampered by a narrow therapeutic window and limited response rate. Two such cytokines are interleukin 2 (IL-2) and interleukin 12 (IL-12), which potently synergize to activate and proliferate T cells and natural killer (NK) cells. However, the only approved human IL-2 therapy, Proleukin, is rarely used in the clinic due to systemic toxicities, and no IL-12 product has been approved to date due to severe dose-limiting toxicities. Here, we describe CLN-617, a first-in-class therapeutic for intratumoral (IT) injection that co-delivers IL-2 and IL-12 on a single molecule in a safe and effective manner. CLN-617 is a single-chain fusion protein comprised of IL-2, leukocyte-associated immunoglobulin-like receptor 2 (LAIR2), human serum albumin (HSA), and IL-12. LAIR2 and HSA function to retain CLN-617 in the treated tumor by binding collagen and increasing molecular weight, respectively. IT administration of a murine surrogate of CLN-617, mCLN-617, eradicates established treated and untreated tumors in syngeneic models, synergizes with anti-PD1 therapy, and generates a robust abscopal response dependent on cellular immunity and antigen cross-presentation. CLN-617 is being evaluated in a clinical trial in patients with advanced solid tumors (NCT06035744).","prettyUrl":"mehta-2024-cir","following":false,"created":"04/02/2024","featured":false,"publishedDate":"04/03/2024","urlOrId":"mehta-2024-cir","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"4cc41219-0c45-4db4-afc2-b7266b16855b","title":"Third-Party Cytotoxic T Lymphocytes for High-Risk Patients with Covid-19","investigator":"Grosso, Dolores","investigatorInstitution":"Thomas Jefferson University Sidney Kimmel Cancer Center, Department of Medical Oncology","publicationName":"TBD","researchArea":"Infectious Disease","prettyUrls":{"325":"grosso-2023-nc"},"prettyUrlList":["grosso-2023-nc"],"summary":"Treatment with off-the-shelf cellular therapy may provide direct and rapid treatment for COVID-19, overcoming the delayed adaptive immune responses associated with poor outcomes in high-risk patients. Thirty ambulatory patients with COVID-19 were enrolled on a phase I trial to assess the safety of 3rd party, COVID-19-specific cytotoxic T lymphocytes (CTLs). Twelve “Interventional” patients matching the HLA-A*02:01 restriction of the CTLs received a single infusion of one of four escalating doses of a product containing 68.5% COVID-19-specific CD8+ CTLs/total cells. Eighteen “Observational” patients lacking HLA-A*02:01 served as comparisons. No dose-limiting toxicities were observed. Nasal swab PCR data showed ≥ 88% viral elimination in 92% of patients in 4 days and the CTLs remained detectable at 6 months. Interventional patients consistently reported symptomatic improvement 2-3 days after infusion, whereas improvement was more variable in Observational patients. Our study shows that COVID-19-specific CTLs are a potentially useful cellular therapy for COVID-19.","prettyUrl":"grosso-2023-nc","following":false,"created":"03/26/2024","featured":false,"publishedDate":"03/26/2024","urlOrId":"grosso-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"0a424f8e-c83e-463e-9beb-a38c3db17ffe","title":"Personalized Neoantigen Vaccine and Pembrolizumab in Advanced Hepatocellular Carcinoma: A phase 1/2 trial","investigator":"Yarchoan, Mark","investigatorInstitution":"Bloomberg–Kimmel Institute for Cancer Immunotherapy. Johns Hopkins University School of Medicine","publicationName":"Nature Medicine","researchArea":"Cancer","prettyUrls":{"323":"yarchoan-2024-nm"},"prettyUrlList":["yarchoan-2024-nm"],"summary":"PD-1 inhibitors have modest efficacy as monotherapy in hepatocellular carcinoma (HCC). A personalized therapeutic cancer vaccine (PTCV) may enhance responses to PD-1 inhibitors through the induction of tumor-specific immunity. We present results of a single-arm, open-label, phase 1/2 study (NCT04251117) of a DNA plasmid PTCV (GNOS-PV02) encoding up to 40 neoantigens co-administered with plasmid-encoded IL-12, plus pembrolizumab, in advanced HCC patients previously treated with a multi-tyrosine kinase inhibitor (mTKI). The most common treatment-related adverse events were injection site reactions, observed in 41.6% (15/36) of patient. No dose-limiting toxicities or treatment-related grade ³3 events were observed. ORR (mITT) per RECIST 1.1 was 30.6% (11/36) with 8.3% (3/36) of patients achieving a complete response (CR). Clinical responses were associated with the number of neoantigens encoded in the vaccine. Neoantigen-specific T cell responses were confirmed in 19/22 (86.4%) evaluable patients by ELISpot. Multi-parametric cellular profiling revealed active, proliferative, and cytolytic vaccine-specific CD4+ and CD8+ effector T cells. In 14/14 (100%) of patients with paired pre- and on-treatment blood and tumor biopsies, we identified by TCRβ bulk sequencing expanded T cells clones in the peripheral blood that also trafficked into the tumor. Single-cell analysis revealed post-treatment T cell clonal expansion of cytotoxic T cell phenotypes. TCR complementarity determining region (CDR) cloning of expanded T cell clones in the tumors post-vaccination confirmed reactivity against vaccine-encoded neoantigens. Our results support the PTCV mechanism of action based on the induction of anti-tumor T cells, and show that a PTCV plus pembrolizumab has clinical activity in advanced HCC.","prettyUrl":"yarchoan-2024-nm","following":false,"created":"02/07/2024","featured":false,"publishedDate":"03/19/2024","urlOrId":"yarchoan-2024-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"3deb5a1d-35ec-4499-840d-629b176571e1","title":"COGNATE ANTIGEN ENGAGEMENT INDUCES HIV-1 EXPRESSION IN CD4+ T CELLS FROM PEOPLE ON LONG-TERM ART","investigator":"Simonetti, F","investigatorInstitution":"Johns Hopkins University, School of Medicine","publicationName":"TBD","researchArea":"HIV","prettyUrls":{"322":"moskovljevic_2024_s"},"prettyUrlList":["moskovljevic_2024_s"],"summary":"TBD","prettyUrl":"moskovljevic_2024_s","following":false,"created":"03/07/2024","featured":false,"publishedDate":"03/07/2024","urlOrId":"moskovljevic_2024_s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"164cfc6a-82bb-4cca-998c-a02ce967c44d","title":"Identification of apolipoprotein B–reactive CDR3 motifs allows tracking of atherosclerosis-related memory CD4+T cells in multiple donors","investigator":"Ley, Klaus","investigatorInstitution":"Immunology Center of Georgia","publicationName":"Frontiers in Immunology","researchArea":"Other","prettyUrls":{"321":"roy-2024-fi"},"prettyUrlList":["roy-2024-fi"],"summary":"Introduction: Atherosclerosis is a major pathological condition that underlies many cardiovascular diseases (CVDs). Its etiology involves breach of tolerance to self, leading to clonal expansion of autoreactive apolipoprotein B (APOB)–reactive CD4+T cells that correlates with clinical CVD. The T-cell receptor (TCR) sequences that mediate activation of APOB-specific CD4+T cells are unknown.\nMethods: In a previous study, we had profiled the hypervariable complementarity determining region 3 (CDR3) of CD4+T cells that respond to six immunodominant APOB epitopes in most donors. Here, we comprehensively analyze this dataset of 149,065 APOB-reactive and 199,211 non-reactive control CDR3s from six human leukocyte antigen–typed donors.\nResults: We identified 672 highly expanded (frequency threshold > 1.39E-03) clones that were significantly enriched in the APOB-reactive group as compared to the controls (log10 odds ratio ≥1, Fisher’s test p < 0.01). Analysis of 114,755 naïve, 91,001 central memory (TCM) and 29,839 effector memory (TEM) CDR3 sequences from the same donors revealed that APOB+ clones can be traced to the complex repertoire of unenriched blood T cells. The fraction of APOB+ clones that overlapped with memory CDR3s ranged from 2.2% to 46% (average 16.4%). This was significantly higher than their overlap with the naïve pool, which ranged from 0.7% to 2% (average 1.36%). CDR3 motif analysis with the machine learning–based\nin-silico tool, GLIPHs (grouping of lymphocyte interactions by paratope hotspots), identified 532 APOB+ motifs. Analysis of naïve and memory CDR3 sequences with GLIPH revealed that ~40% (209 of 532) of these APOB+motifs were enriched in the memory pool. Network analysis with Cytoscape revealed extensive sharing of the\nmemory-affiliated APOB+ motifs across multiple donors. We identified six motifs that were present in TCM and TEM CDR3 sequences from >80% of the donors and were highly enriched in the APOB-reactive TCR repertoire.\nDiscussion: The identified APOB-reactive expanded CD4+T cell clones and conserved motifs can be used to annotate and track human atherosclerosis-related autoreactive CD4+T cells and measure their clonal expansion.","prettyUrl":"roy-2024-fi","following":false,"created":"03/07/2024","featured":false,"publishedDate":"03/07/2024","urlOrId":"roy-2024-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d1877678-b683-4b19-bacd-dcb68ae2af2c","title":"A dietary commensal microbe enhances antitumor immunity by activating tumor macrophages to sequester iron","investigator":"Sharma, G","investigatorInstitution":"Pohang University of Science and Technology (POSTECH), Immunobiome Inc.","publicationName":"Nature Immunology","researchArea":"Other","prettyUrls":{"320":"sharma-2024-ni"},"prettyUrlList":["sharma-2024-ni"],"summary":"Innate immune cells generate a multifaceted antitumor immune response, including the conservation of essential nutrients such as iron. These cells can be modulated by commensal bacteria; however, identifying and understanding how this occurs is a challenge. Here we show that the food commensal Lactiplantibacillus plantarum IMB19 augments antitumor immunity in syngeneic and xenograft mouse tumor models. Its capsular heteropolysaccharide is the major effector molecule, functioning as a ligand for TLR2. In a two-pronged manner, it skews tumor-associated macrophages to a classically active phenotype, leading to generation of a sustained CD8+ T cell response, and triggers macrophage ‘nutritional immunity’ to deploy the high-affinity iron transporter lipocalin-2 for capturing and sequestering iron in the tumor microenvironment. This process induces a cycle of tumor cell death, epitope expansion and subsequent tumor clearance. Together these data indicate that food commensals might be identified and developed into ‘oncobiotics’ for a multi-layered approach to cancer therapy.","prettyUrl":"sharma-2024-ni","following":false,"created":"02/28/2024","featured":false,"publishedDate":"02/28/2024","urlOrId":"sharma-2024-ni","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"93d86797-404f-499e-9c5e-cbcf594d7e9a","title":"Dynamic establishment of recipient resident memory T cell repertoire after human intestinal transplantation","investigator":"Sykes, M","investigatorInstitution":"Columbia University Medical Center","publicationName":"eBioMedicine","researchArea":"Organ transplant","prettyUrls":{"319":"jiao-2024-ebm"},"prettyUrlList":["jiao-2024-ebm"],"summary":"Understanding formation of the human tissue resident memory T cell (TRM) repertoire requires longitudinal access to human non-lymphoid tissues. By applying flow cytometry and next generation sequencing to serial blood, lymphoid tissue, and gut samples from 16 intestinal transplantation (ITx) patients, we assessed the origin, distribution, and specificity of human TRMs at phenotypic and clonal levels. Donor age ≥1 year and blood T cell macrochimerism (peak level ≥4%) were associated with delayed establishment of stable recipient TRM repertoires in the transplanted ileum. T cell receptor (TCR) overlap between paired gut and blood repertoires from ITx patients was significantly greater than that in healthy controls, demonstrating increased gut-blood crosstalk after ITx. Crosstalk with the circulating pool remained high for years of follow-up. TCR sequences identifiable in pre-Tx recipient gut but not those in lymphoid tissues alone were more likely to populate post-Tx ileal allografts. Clones detected in both pre-Tx gut and lymphoid tissue had distinct transcriptional profiles from those identifiable in only one tissue. Recipient T cells were distributed widely throughout the gut, including allograft and native colon, which had substantial repertoire overlap. Both alloreactive and microbe-reactive recipient T cells persisted in transplanted ileum, contributing to the TRM repertoire. Our studies reveal human intestinal TRM repertoire establishment from the circulation, preferentially involving lymphoid tissue counterparts of recipient intestinal T cell clones, including TRMs. We have described the temporal and spatial dynamics affecting this active crosstalk between the circulating pool and the intestinal TRM pool.","prettyUrl":"jiao-2024-ebm","following":false,"created":"02/23/2024","featured":false,"publishedDate":"02/23/2024","urlOrId":"jiao-2024-ebm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a4d73c02-a9eb-4723-9ec0-4714960e2b03","title":"Humans with inherited pre-TCR-⍺ deficiency and ⍺β T cells","investigator":"Béziat, Vivien","investigatorInstitution":"Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children","publicationName":"Science","researchArea":"Immunocompetence","prettyUrls":{"318":"materna-2024-s"},"prettyUrlList":["materna-2024-s"],"summary":"We describe humans with rare biallelic loss-of-function PTCRA variants impairing pre-TCRa expression. Low circulating naïve αβ T cell counts at birth persisted over time, with normal memory αβ and high γδ T cell counts. Their TCRα repertoire was biased, suggesting that noncanonical thymic differentiation pathways can rescue αβ T cell development. Only a minority of these individuals were sick, with infection, lymphoproliferation, and/or autoimmunity. We also report that 1 in 4000 individuals from the Middle East and South Asia are homozygous for a common hypomorphic PTCRA variant. They had normal circulating naïve αβ T cell counts but high γδ T cell counts. Although residual pre-TCRa expression drove the differentiation of more αβ T cells, autoimmune conditions were more frequent in these patients than in the general population.","prettyUrl":"materna-2024-s","following":false,"created":"01/16/2024","featured":false,"publishedDate":"02/21/2024","urlOrId":"materna-2024-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"50048097-6c99-48cc-a7cf-538c3428ae9b","title":"Cancer-specific CD8 T cell frequency at baseline in blood correlates with response to PD-1 blockade in Merkel cell carcinoma","investigator":"Pulliam, Thomas","investigatorInstitution":"University of Washington","publicationName":"","researchArea":"Cancer Immunotherapy","prettyUrls":{"317":"pulliam-2024-crm"},"prettyUrlList":["pulliam-2024-crm"],"summary":"Understanding immunotherapy response and resistance is challenging due to difficulty identifying cancer-specific CD8 T cells. Merkel cell carcinoma (MCC) is typically driven by Merkel cell polyomavirus (MCPyV), facilitating identification of cancer-specific T cells across patients. We characterized cancer-specific T cells in 35 MCC patients, including from a neoadjuvant anti-PD-1 trial. Higher MCPyV-specific CD8 T-cell frequency in pre-treatment blood (but not tumors) correlated with response (p=0.0056). Single cell RNAseq revealed MCPyV-specific CD8 T cells in blood with increased stem/memory signatures and decreased exhaustion signatures relative to their intratumoral counterparts. The number of circulating cancer-specific T cells appears most linked to initial response to immunotherapy. Indeed, a longitudinal study showed loss of MHC-I on tumor cells during newly acquired resistance despite abundant cancer-specific CD8 T cells in blood. (original) These results suggest that blood acts as an important reservoir of cancer-specific CD8 T cells and suggests adoptive cell therapies may be particularly effective in patients without such cells.","prettyUrl":"pulliam-2024-crm","following":false,"created":"03/10/2023","featured":false,"publishedDate":"02/08/2024","urlOrId":"pulliam-2024-crm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"14e895c9-fdf5-4704-9945-c11ac03f737f","title":"The signature of a T-cell response to KSHV persists across space and time in individuals with epidemic and endemic KS from Uganda","investigator":"Edus H. Warren","investigatorInstitution":"Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center","publicationName":"","researchArea":"Cancer","prettyUrls":{"316":"ravishankar-2024-jem"},"prettyUrlList":["ravishankar-2024-jem"],"summary":"Coming Soon...","prettyUrl":"ravishankar-2024-jem","following":false,"created":"11/08/2023","featured":false,"publishedDate":"01/29/2024","urlOrId":"ravishankar-2024-jem","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"89794211-d4aa-45b5-a5f2-0ef205509a79","title":"Dynamics and survival associations of T cell receptor clusters in patients with pleural mesothelioma treated with immunotherapy","investigator":"Mansfield, Aaron","investigatorInstitution":"Division of Medical Oncology, Mayo Clinic","publicationName":"Journal for ImmunoTherapy of Cancer","researchArea":"Cancer Immunotherapy","prettyUrls":{"315":"desai-2023-jitc"},"prettyUrlList":["desai-2023-jitc"],"summary":"Background: Immune checkpoint inhibitors (ICIs) are now a first-line treatment option for patients with pleural mesothelioma with the recent approval of ipilimumab and nivolumab. Mesothelioma has a low tumor mutation burden and no robust predictors of survival with ICI. Since ICIs enable adaptive antitumor immune responses, we investigated T-cell receptor (TCR) associations with survival in participants from two clinical trials treated with ICI.\n\nMethods: We included patients with pleural mesothelioma who were treated with nivolumab (NivoMes, NCT02497508) or nivolumab and ipilimumab (INITIATE, NCT03048474) after first-line therapy. TCR sequencing was performed with the ImmunoSEQ assay in 49 and 39 pretreatment and post-treatment patient peripheral\nblood mononuclear cell (PBMC) samples. These data were integrated with TCR sequences found in bulk RNAseq data by TRUST4 program in 45 and 35 pretreatment and post-treatment tumor biopsy samples and TCR sequences from over 600 healthy controls. The TCR sequences were clustered into groups of shared antigen specificity using GIANA. Associations of TCR clusters with overall survival were determined by cox proportional hazard analysis. Results We identified 4.2 million and 12 thousand\ncomplementarity-determining region 3 (CDR3) sequences from PBMCs and tumors, respectively, in patients treated with ICI. These CDR3 sequences were integrated with\n2.1 million publically available CDR3 sequences from healthy controls and clustered. ICI-enhanced T-cell infiltration and expanded T cell diversity in tumors. Cases with TCR clones in the top tertile in the pretreatment tissue or in circulation had significantly better survival than the bottom two tertiles (p<0.04). Furthermore, a\nhigh number of shared TCR clones between pretreatment tissue and in circulation was associated with improved survival (p=0.01). To potentially select antitumor clusters, we filtered for clusters that were (1) not found in healthy controls, (2) recurrent in multiple patients with mesothelioma, and (3) more prevalent in post treatment than pretreatment samples. The detection of two-specific TCR clusters provided significant survival benefit compared with detection of 1 cluster (HR<0.001, p=0.026) or the detection of no TCR clusters (HR=0.10, p=0.002). These two clusters were not found in bulk tissue RNA-seq data and have not been reported in public CDR3 databases.\n\nConclusions: We identified two unique TCR clusters that were associated with survival on treatment with ICI in patients with pleural mesothelioma. These clusters may enable approaches for antigen discovery and inform future targets for design of adoptive T cell therapies.","prettyUrl":"desai-2023-jitc","following":false,"created":"06/06/2023","featured":false,"publishedDate":"01/04/2024","urlOrId":"desai-2023-jitc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b9107078-7d40-44ff-9ae6-1f26c65dff73","title":"Perturbations of the T-cell receptor repertoire in response to SARS-CoV-2 in immunocompetent and immunocompromised individuals","investigator":"Notarangelo, Luigi","investigatorInstitution":"Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health","publicationName":"Journal of Allergy and Clinical Immunology","researchArea":"Infectious Disease","prettyUrls":{"314":"delmonte-2023-jaci"},"prettyUrlList":["delmonte-2023-jaci"],"summary":"Background: Functional T-cell responses are essential for virus clearance and long-term protection after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas certain clinical factors, such as older age and immunocompromise, are associated with worse outcome.\n\nObjectives: We studied the breadth and magnitude of the T-cell responses in COVID-19 patients and in individuals with inborn errors of immunity (IEI) who had received COVID-19 mRNA vaccine.\n\nMethods: Utilizing high-throughput sequencing and bioinformatics tools to characterize the T-cell receptor β (TRB) repertoire signatures in 540 individuals after SARS-CoV-2 infection, 31 IEI recipients of COVID-19 mRNA vaccine, and in healthy controls, we quantified HLA class-I- and class II-restricted SARS-CoV-2-specific responses and also identified several HLA allele-clonotype motif associations in COVID-19 patients, including a sub-cohort of anti-type I interferon (IFN-I)-positive patients.\n\nResults: Our analysis revealed that elderly COVID-19 patients with critical disease manifested lower SARS-CoV-2 T-cell clonotype diversity as well as T-cell responses with reduced magnitude, whereas the SARS-CoV-2-specific clonotypes targeted a broad range of HLA class I- and class-II-restricted epitopes across the viral proteome. The presence of anti-IFN-I antibodies was associated with certain HLA alleles. Finally, COVID-19 mRNA immunization induced an increase in the breadth of SARS-CoV-2-specific clonotypes in patients with IEI, including those who had failed to seroconvert.\n\nConclusions: Elderly individuals have impaired capacity to develop broad and sustained T-cell responses after SARS-CoV-2 infection. Genetic factors may play a role in the production of anti-IFN-I antibodies. COVID-19 mRNA vaccines are effective in inducing T-cell responses in patients with IEI.","prettyUrl":"delmonte-2023-jaci","following":false,"created":"04/19/2023","featured":false,"publishedDate":"01/04/2024","urlOrId":"delmonte-2023-jaci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b07139f8-baa4-48ae-abd4-1ed751396b80","title":"Unstable regulatory and autoreactive effector T cells in patients with FOXP3 mutations","investigator":"Borna, Šimon","investigatorInstitution":"Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine","publicationName":"Science Translational Medicine","researchArea":"Basic Immunology","prettyUrls":{"313":"borna-2023-fi"},"prettyUrlList":["borna-2023-fi"],"summary":"Studies of the monogenic autoimmune disease immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX) have elucidated the essential function of the transcription factor FOXP3 and thymic-derived regulatory T cells (Tregs) in controlling peripheral tolerance. However, the presence and the source of autoreactive T cells in IPEX remain undetermined. Here, we investigated how FOXP3 deficiency affects the T cell receptor (TCR) repertoire and Treg stability in vivo and compared T cell abnormalities in patients with IPEX to those in patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy syndrome (APECED). To study Tregs independently of their phenotype and to analyze T cell autoreactivity, we combined Treg-specific demethylation region analyses, single-cell multi-omic profiling, and bulk TCR sequencing. We found that patients with IPEX, unlike patients with APECED, have expanded autoreactive T cells originating from both autoreactive effector T cells (Teffs) and Tregs. In addition, a fraction of the expanded Tregs from patients with IPEX lost their phenotypic and functional markers including CD25 and FOXP3. Functional experiments with CRISPR/Cas9-mediated FOXP3 knock-out Tregs and Tregs from patients with IPEX indicated that the patients’ Tregs gain a Th2 skewed Teff-like function, which is consistent with immune dysregulation observed in these patients. Analyses of FOXP3 mutation-carrier mothers and a patient with IPEX after hematopoietic stem cell transplantation, indicated that Tregs expressing non-mutated FOXP3 prevent the accumulation of autoreactive Teffs and unstable Tregs. These findings could be directly used for diagnostic and prognostic purposes and for monitoring the effects of immunomodulatory treatments.","prettyUrl":"borna-2023-fi","following":false,"created":"11/06/2023","featured":false,"publishedDate":"12/20/2023","urlOrId":"borna-2023-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"2c853ddc-1f30-4134-8c7d-cdd9db5a3a61","title":"Autoreactive T cells target peripheral nerves in Guillain–Barré syndrome","investigator":"Latorre, Daniela","investigatorInstitution":"ETH Zurich, Institute of Microbiology, Human Neuroimmunology Group","publicationName":"Nature","researchArea":"Autoimmune Disorders","prettyUrls":{"311":"sukenikova-2023-n"},"prettyUrlList":["sukenikova-2023-n"],"summary":"Guillain-Barré syndrome (GBS) is a rare heterogenous disorder of the peripheral nervous system, which is usually triggered by a preceding infection, and causes a potentially life-threatening progressive muscle weakness1. Although GBS is considered an autoimmune disease, the mechanisms that underlie its distinct clinical subtypes remain largely unknown. Here, by combining in vitro T cell screening, single-cell RNA sequencing and T cell receptor (TCR) sequencing, we identify autoreactive memory CD4+ cells, that show a cytotoxic T helper 1 (TH1)-like phenotype, and rare CD8+ T cells that target myelin antigens of the peripheral nerves in patients with the demyelinating disease variant. We characterized more than 1,000 autoreactive single T cell clones, which revealed a polyclonal TCR repertoire, short CDR3β lengths, preferential HLA-DR restrictions and recognition of immunodominant epitopes. We found that autoreactive TCRβ clonotypes were expanded in the blood of the same patient at distinct disease stages and, notably, that they were shared in the blood and the cerebrospinal fluid across different patients with GBS, but not in control individuals. Finally, we identified myelin-reactive T cells in the nerve biopsy from one patient, which indicates that these cells contribute directly to disease pathophysiology. Collectively, our data provide clear evidence of autoreactive T cell immunity in a subset of patients with GBS, and open new perspectives in the field of inflammatory peripheral neuropathies, with potential impact for biomedical applications.","prettyUrl":"sukenikova-2023-n","following":false,"created":"10/19/2023","featured":false,"publishedDate":"12/19/2023","urlOrId":"sukenikova-2023-n","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"f4d7fe41-ce2d-4865-96b4-41e44fd2f87e","title":"TCR sequencing of HBRV stimulated cells, Crohn's blood, lymph node and colon, and blood and lymph node in primary biliary cholangitis","investigator":"Mason, Andrew","investigatorInstitution":"University of Alberta, Department of Medicine","publicationName":"","researchArea":"Basic Immunology","prettyUrls":{"310":"mason-2023"},"prettyUrlList":["mason-2023"],"summary":"HBRV stilumated cells: Intrahepatic lymphocytes obtained by flushing the liver of Primary Biliary Cholangitis patients undergoing transplant were stimulated with Env (E) and Gag (G) peptides from the Human Betaretrovirus (HBRV) and activated CD8+ CD137+ and CD4+ CD154+ were sorted and sequenced.\n\nCrohn’s: Samples obtained from pediatric and adult Crohn's disease patients were sequenced for their TCR. TP-C identifies Colon Tissue samples from pediatric patients, TP-LN identifies Lymph Node samples from pediatric patients, GR identifies whole blood samples from adult Crohn's disease patients and CD-LN identifies lymph node samples obtained from adult Crohn's disease patients.\n\nPrimary Biliary Cholangitis: TCR sequences obtained from Intrahepatic Lymphocyte (IHL) whole blood, Lymph nodes (LN), Whole Blood (RV) and Sorted CD4+ and CD8+ cells from whole blood (RV-CD4 or CD8). Serial RV samples with years indicating year of collection are also indicated in the sample ID.","prettyUrl":"mason-2023","following":false,"created":"11/28/2023","featured":false,"publishedDate":"12/06/2023","urlOrId":"mason-2023","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"11a66867-470a-4672-a63f-a30d0a59ece0","title":"Plasticity of intragraft alloreactive T cell clones in human gut correlates with transplant outcomes","investigator":"Sykes, Megan","investigatorInstitution":"Columbia Center for Translational Immunology, Department of Medicine, Columbia University","publicationName":"Journal of Experimental Medicine","researchArea":"Organ transplant","prettyUrls":{"309":"fu-2023-jem"},"prettyUrlList":["fu-2023-jem"],"summary":"The site of transition between tissue resident memory (TRM) and circulating phenotypes of T cells is unknown. We integrated clonotype, alloreactivity and gene expression profiles of graft-repopulating recipient T cells in the intestinal mucosa at the single cell level after human intestinal transplantation. Host-versus-graft (HvG)-reactive T cells mainly distributed to TRM, Teff/TRM and T follicular helper compartments. RNA velocity analysis demonstrated a trajectory from TRM to effector T (Teff)/TRM clusters in association with rejection. By integrating pre- and post-Tx mixed lymphocyte reaction-determined alloreactive repertoires, we observed that pre-existing HvG-reactive T cells that demonstrated tolerance in the circulation were dominated by TRM profiles in quiescent allografts. Putative de novo HvG-reactive clones showed a transcriptional profile skewed to cytotoxic effectors in rejecting grafts. Inferred protein regulon network analysis revealed upstream regulators that accounted for the effector and tolerant T cell states. We demonstrate Teff/TRM interchangeability for individual T cell clones with known (allo)recognition in human gut, providing novel insight into TRM biology.","prettyUrl":"fu-2023-jem","following":false,"created":"02/13/2023","featured":false,"publishedDate":"11/20/2023","urlOrId":"fu-2023-jem","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"55029ec5-db82-48b2-b1d6-3e2acbc4c96f","title":"Tumor reactive γδ T cells contribute to a complete response to PD-1 blockade in a Merkel cell carcinoma patient","investigator":"Lien, Scott","investigatorInstitution":"Department of Immunology, University of Toronto, Toronto","publicationName":"Nature Communications","researchArea":"Cancer Immunotherapy","prettyUrls":{"308":"lien-2023-nc"},"prettyUrlList":["lien-2023-nc"],"summary":"Immunotherapies targeting PD-1/PD-L1 are now widely used in the clinic to treat a variety of malignancies. While the majority of research on T cell exhaustion and PD-1 blockade has been focused on conventional αβ T cells, the contribution of innate-like T cells such as γδ T cells to anti-PD-1/PD-L1 mediated therapy is limited. Here we show that tumor reactive γδ T cells respond to PD-1 blockade in a Merkel cell carcinoma (MCC) patient experiencing a complete response to therapy. We find clonally expanded γδ T cells in the blood and tumor after pembrolizumab treatment, and this Vγ2Vδ1 clonotype recognizes Merkel cancer cells in a TCR-dependent manner. Notably, the intra-tumoral γδ T cells in the MCC patient are characterized by higher expression of PD-1 and TIGIT, relative to conventional CD4 and CD8 T cells. Our results demonstrate that innate-like T cells could also contribute to an anti-tumor response after PD-1 blockade.","prettyUrl":"lien-2023-nc","following":false,"created":"11/06/2023","featured":false,"publishedDate":"11/14/2023","urlOrId":"lien-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"71696fdf-c819-4a8f-b7c2-57646c03b25d","title":"Comprehensive immune profiling of SARS-CoV-2 infected kidney transplant patients","investigator":"Fenninger, Franz","investigatorInstitution":"Department of Medicine, University of British Columbia","publicationName":"","researchArea":"Organ transplant","prettyUrls":{"307":"fenninger-2023-ft"},"prettyUrlList":["fenninger-2023-ft"],"summary":"The immune responses of kidney transplant recipients against SARS-CoV-2 remains under studied. In this prospective pilot study, we performed comprehensive immune profiling using cellular, proteomic, and serologic assays on a cohort of 9 kidney transplant recipients and 12 non-transplant individuals diagnosed with COVID-19. Our data show that in addition to having reduced SARS-CoV-2 specific antibody levels, kidney transplant recipients exhibited significant cellular differences including a decrease in naïve - but increase in effector T cells, a high number of CD28+ CD4 effector memory T cells, and increased CD8 T memory stem cells compared with non-transplant patients. Furthermore, transplant patients had lower concentrations of serum cytokine MIP-1β as well as a less diverse T cell receptor repertoire. Overall, our results show that compared to non-transplant patients, kidney transplant recipients with SARS-CoV-2 infection exhibit an immunophenotype that is reminiscent of the immune signature observed in patients with severe COVID-19.","prettyUrl":"fenninger-2023-ft","following":false,"created":"10/18/2023","featured":false,"publishedDate":"10/19/2023","urlOrId":"fenninger-2023-ft","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"44a0d585-1966-47db-9e32-8919277d07bf","title":"Neoepitope-specific vaccination of patients with diffuse midline glioma targeting H3K27M induces polyclonal B and T cell responses across diverse HLA alleles","investigator":"Boschert, Tamara","investigatorInstitution":"CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ) /Faculty of Biosciences, Heidelberg University/Helmholtz Institute for Translational Oncology (HI-TRON) & BioMed X GmbH","publicationName":"BioRxiv","researchArea":"Cancer Immunotherapy","prettyUrls":{"306":"boschert-2023-nm"},"prettyUrlList":["boschert-2023-nm"],"summary":"Coming Soon...","prettyUrl":"boschert-2023-nm","following":false,"created":"04/05/2023","featured":false,"publishedDate":"10/13/2023","urlOrId":"boschert-2023-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"743cd8c2-6d03-4448-a46e-13fae4487ebd","title":"Fc-enhanced anti-CTLA-4 antibody effects on T cell compartment","investigator":"Chloe Delepine","investigatorInstitution":"Agenus, Inc.","publicationName":"","researchArea":"Cancer Immunotherapy","prettyUrls":{"305":"delepine-2023-nm"},"prettyUrlList":["delepine-2023-nm"],"summary":"Conventional immune checkpoint inhibitors (ICI) targeting CTLA-4 elicit durable survival, but primarily in patients with immune-inflamed tumors. Although the mechanisms underlying response to anti-CTLA-4 remain poorly understood, Fc-gamma receptor (FcγR) IIIA co engagement appears critical for activity, potentially explaining the modest clinical benefits of approved anti-CTLA-4 antibodies. We demonstrate that anti-CTLA-4 engineered for enhanced FcγR affinity leverages FcγR-dependent mechanisms to potentiate T cell responsiveness, reduce intratumoral Tregs, and enhance antigen presenting cell activation. Fc-enhanced anti-CTLA-4 promoted superior efficacy in mouse models and remodeled innate and adaptive immunity versus conventional anti-CTLA-4. These findings extend to patients treated with botensilimab, an Fc-enhanced anti-CTLA-4 antibody, with clinical activity across multiple poorly immunogenic and ICI treatment-refractory cancers. Efficacy was independent of tumor neoantigen burden or FcγRIIIA genotype. However, FcγRIIA and FcγRIIIA expression emerged as potential response biomarkers. These data highlight the therapeutic potential of Fc-enhanced anti-CTLA-4 antibodies in cancers unresponsive to conventional ICI therapy.","prettyUrl":"delepine-2023-nm","following":false,"created":"09/20/2023","featured":false,"publishedDate":"09/20/2023","urlOrId":"delepine-2023-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"8f43f5d4-6f02-4121-960a-10bb5f8d67b0","title":"Tumor-resident Lactobacillus iners confers chemoradiation resistance through lactate-induced metabolic rewiring","investigator":"Colbert, Lauren E.","investigatorInstitution":"The University of Texas M.D. Anderson Cancer Center","publicationName":"Cancer Cell","researchArea":"Cancer","prettyUrls":{"303":"colbert-2023-cc"},"prettyUrlList":["colbert-2023-cc"],"summary":"Tumor microbiota can produce active metabolites that affect cancer and immune cell signaling, metabolism, and proliferation. Here, we explore tumor and gut microbiome features that affect chemoradiation response in cervical cancer patients through a combined approach of deep microbiome sequencing, sequential, targeted bacterial culture and in vitro assays. We identify that an obligate L-lactate producing lactic acid bacteria found in tumors, Lactobacillus iners, is associated with decreased survival in patients, induces chemotherapy and radiation resistance in cervical cancer cells, and leads to metabolic rewiring of tumors. Genomically similar L-lactate producing lactic acid bacteria commensal to other body sites are also significantly associated with survival in colorectal cancer, lung cancer, head and neck cancer and skin cancer. Our findings demonstrate that lactic acid bacteria in the tumor microenvironment can alter tumor metabolism and lactate signaling pathways to cause therapy resistance. Lactic acid bacteria could be promising therapeutic targets across cancer types.","prettyUrl":"colbert-2023-cc","following":false,"created":"09/05/2023","featured":false,"publishedDate":"09/07/2023","urlOrId":"colbert-2023-cc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"49a31e80-cc36-41f9-a915-d95835c3324e","title":"NL-201 Upregulates MHC-I Expression and Intratumoral T-cell Receptor Diversity, and Demonstrates Robust Antitumor Activity as Monotherapy and in Combination with PD-1 Blockade","investigator":"Huard, Justin","investigatorInstitution":"Neoleukin Therapuetics","publicationName":"Cancer Immunology Research","researchArea":"Cancer Immunotherapy","prettyUrls":{"302":"cm-2023-cir"},"prettyUrlList":["cm-2023-cir"],"summary":"Cytokine engineering has shown promise as a means to create novel immunomodulatory agents or to improve upon the therapeutic potential of natural cytokines. NL-201, a de novo, hyperstable, IL2 receptor alpha (IL2Rα)-independent agonist of the receptors for IL2 and IL15, elicits robust preclinical activity in syngeneic murine cancer models, including those resistant to immune checkpoint inhibitors (ICI). Here, we report that NL-201 monotherapy converts 'cold' tumor microenvironments (TME) to immunologically 'hot' states by driving pro-inflammatory gene expression, enhancing IFNγ-dependent MHC-I expression, and expanding both T-cell number and clonal diversity. In addition, the combination of NL-201 and anti-PD-1 resulted in complementary antitumor activity in the immunologically 'cold' and ICI resistant B16F10, EMT6, and Renca syngeneic models. In the B16F10 model, treatment with NL-201 plus anti-PD-1 increased the abundance of CD4+ and CD8+ effector T cells in the TME. These findings reveal an important mechanistic basis for the antitumor activity of NL-201 both as a monotherapy and in combination with PD-1 antagonists, and provide further context for the role of IL2Rα-based signaling in ICI-resistant tumors.","prettyUrl":"cm-2023-cir","following":false,"created":"06/30/2023","featured":false,"publishedDate":"08/28/2023","urlOrId":"cm-2023-cir","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a7bf2c51-6fca-420b-a275-17a05831b725","title":"CDK4/6 inhibition sensitizes intracranial tumors to PD-1 blockade in preclinical models of brain metastasis","investigator":"Brastianos, Priscilla","investigatorInstitution":"Massachusetts General Hospital","publicationName":"Clinical Cancer Research","researchArea":"Cancer Immunotherapy","prettyUrls":{"301":"nayyar-2023-ccr"},"prettyUrlList":["nayyar-2023-ccr"],"summary":"Purpose: Brain metastases are associated with high morbidity and often resistant to immune checkpoint inhibitors. We evaluated whether CDK4/6 inhibitor (CDKi) abemaciclib can sensitize intracranial tumors to PD-1 inhibition in mouse models of melanoma and breast cancer brain metastasis.\n\nExperimental Design: Treatment response was evaluated in vivo using immunocompetent mouse models of brain metastasis bearing concurrent intracranial and extracranial tumors. Treatment effect on intracranial and extracranial tumor immune microenvironments was evaluated using immunofluorescence, multiplex immunoassays, high-parameter flow cytometry and T cell receptor profiling. Mice with humanized immune systems were evaluated using flow cytometry to study the effect of CDKi on human T cell development.\n\nResults: We found that combining abemaciclib with PD-1 inhibition reduced tumor burden and improved overall survival in mice. The tumor immune microenvironment, which differed based on anatomical location of tumors, was altered with CDKi and PD-1 inhibition in an organ-specific manner. Combination abemaciclib and anti-PD-1 treatment increased recruitment and expansion of CD8+ effector T cell subsets, depleted CD4+ regulatory T (TREG) cells, and reduced levels of immunosuppressive cytokines in intracranial tumors. In immunodeficient mice engrafted with human immune systems, abemaciclib treatment supported development and maintenance of CD8+ T cells and depleted TREG cells.\n\nConclusions: Our results highlight the distinct properties of intracranial and extracranial tumors and support clinical investigation of combination CDK4/6 and PD-1 inhibition in patients with brain metastases.","prettyUrl":"nayyar-2023-ccr","following":false,"created":"08/03/2023","featured":false,"publishedDate":"08/07/2023","urlOrId":"nayyar-2023-ccr","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a2e2a003-d454-48b0-b024-bd31bd655c72","title":"Low immune infiltrate level is associated with worse overall survival and complete loss of TP53 and RB1 in pleomorphic rhabdomyosarcoma","investigator":"Futreal, P Anderw","investigatorInstitution":"Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center,","publicationName":"Human Genetics and Genomics Advances","researchArea":"Cancer","prettyUrls":{"300":"beird-2023-hgga"},"prettyUrlList":["beird-2023-hgga"],"summary":"Rhabdomyosarcoma accounts for roughly 1% of adult sarcomas, with pleomorphic rhabdomyosarcoma (PRMS) as the most common subtype. Survival outcomes remain poor for patients with PRMS and little is known about the molecular drivers of this disease. To better characterize PRMS, we performed a broad array of genomic and immunostaining analyses on 25 patient samples. In terms of gene expression and methylation, PRMS clustered more closely with other complex karyotype sarcomas than with pediatric alveolar and embryonal rhabdomyosarcoma. Immune infiltrate levels in PRMS were among the highest observed in multiple sarcoma types and contrasted with low levels in other rhabdomyosarcoma subtypes. Higher immune infiltrate levels were associated with improved overall survival while lower immune infiltrate was associated with complete loss of both TP53 and RB1. This comprehensive characterization of the genetic, epigenetic and immune landscape of PRMS provides a roadmap for improved prognostications and therapeutic exploration.","prettyUrl":"beird-2023-hgga","following":false,"created":"07/28/2023","featured":false,"publishedDate":"08/02/2023","urlOrId":"beird-2023-hgga","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"42f65f69-9967-468b-b6dd-82971f9c9d92","title":"Immunotherapy targeting different immune compartments in combination with radiation therapy induces regression of resistant tumors","investigator":"Rudqvist, Nils-Petter","investigatorInstitution":"Weill Cornell Medicine","publicationName":"Nature Communications","researchArea":"Cancer Immunotherapy","prettyUrls":{"299":"rudqvist-2023-nc"},"prettyUrlList":["rudqvist-2023-nc"],"summary":"Radiation therapy (RT) increases tumor response to CTLA-4 inhibition (CTLA4i) in mice and in some patients, yet deep responses are rare. To identify rational combinations of immunotherapy to improve responses we use models of triple negative breast cancer highly resistant to immunotherapy in female mice. We find that CTLA4i promotes the expansion of CD4+ T helper cells, whereas RT enhances T cell clonality and enriches for CD8+ T cells with an exhausted phenotype. Combination therapy decreases regulatory CD4+ T cells and increases effector memory, early activation and precursor exhausted CD8+ T cells. A combined gene signature comprising these three CD8+ T cell clusters is associated with survival in patients. Here we show that targeting additional immune checkpoints expressed by intratumoral T cells, including PD1, is not effective, whereas CD40 agonist therapy recruits resistant tumors into responding to the combination of RT and CTLA4i, indicating the need to target different immune compartments.","prettyUrl":"rudqvist-2023-nc","following":false,"created":"07/31/2023","featured":false,"publishedDate":"08/02/2023","urlOrId":"rudqvist-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"fa503086-4201-46bb-adcf-be765e1830eb","title":"Immunologic Characterization and T cell Receptor Repertoires of Expanded Tumor-infiltrating Lymphocytes in Patients with Renal Cell Carcinoma","investigator":"Mustjoki, Satu","investigatorInstitution":"Hematology Research Unit, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center","publicationName":"Cancer Research Communications","researchArea":"Cancer","prettyUrls":{"298":"lee-2023-crc"},"prettyUrlList":["lee-2023-crc"],"summary":"The successful use of expanded tumor-infiltrating lymphocytes (TIL) in adoptive TIL therapies has been reported, but the effects of the TIL expansion, immunophenotype, function, and T cell receptor (TCR) repertoire of the infused products relative to the tumor microenvironment (TME) are not well understood. In this study, we analyzed the tumor samples (n = 58) from treatment-naïve patients with renal cell carcinoma (RCC), \"pre-rapidly expanded\" TILs (pre-REP TIL, n = 15) and \"rapidly expanded\" TILs (REP TIL, n = 25) according to a clinical-grade TIL production protocol, with single-cell RNA (scRNA)+TCRαβ-seq (TCRαβ sequencing), TCRβ-sequencing (TCRβ-seq), and flow cytometry. REP TILs encompassed a greater abundance of CD4+ than CD8+ T cells, with increased LAG-3 and low PD-1 expressions in both CD4+ and CD8+ T cell compartments compared with the pre-REP TIL and tumor T cells. The REP protocol preferentially expanded small clones of the CD4+ phenotype (CD4, IL7R, KLRB1) in the TME, indicating that the largest exhausted T cell clones in the tumor do not expand during the expansion protocol. In addition, by generating a catalog of RCC-associated TCR motifs from >1,000 scRNA+TCRαβ-seq and TCRβ-seq RCC, healthy and other cancer sample cohorts, we quantified the RCC-associated TCRs from the expansion protocol. Unlike the low-remaining amount of anti-viral TCRs throughout the expansion, the quantity of the RCC-associated TCRs was high in the tumors and pre-REP TILs but decreased in the REP TILs. Our results provide an in-depth understanding of the origin, phenotype, and TCR specificity of RCC TIL products, paving the way for a more rationalized production of TILs.","prettyUrl":"lee-2023-crc","following":false,"created":"03/27/2023","featured":false,"publishedDate":"07/24/2023","urlOrId":"lee-2023-crc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"ee5df40f-f728-4731-bb7c-d52f8aaa3505","title":"Hedgehog Costimulation During Ischemia Reperfusion Injury Potentiates Cytokine and Homing Responses of CD4+ T Cells","investigator":"Jane-wit, Dan","investigatorInstitution":"Yale University","publicationName":"Frontiers in Immunology","researchArea":"Other","prettyUrls":{"297":"wang-2023-fi"},"prettyUrlList":["wang-2023-fi"],"summary":"Introduction: Ischemia reperfusion injury (IRI) confers worsened outcomes and is an increasing clinical problem in solid organ transplantation. Previously, we identified a \"PtchHi\" T-cell subset that selectively received costimulatory signals from endothelial cell-derived Hedgehog (Hh) morphogens to mediate IRI-induced vascular inflammation.\n\nMethods: Here, we used multi-omics approaches and developed a humanized mouse model to resolve functional and migratory heterogeneity within the PtchHi population.\n\nResults: Hh-mediated costimulation induced oligoclonal and polyclonal expansion of clones within the PtchHi population, and we visualized three distinct subsets within inflamed, IRI-treated human skin xenografts exhibiting polyfunctional cytokine responses. One of these PtchHi subsets displayed features resembling recently described T peripheral helper cells, including elaboration of IFN-y and IL-21, expression of ICOS and PD-1, and upregulation of positioning molecules conferring recruitment and retention within peripheral but not lymphoid tissues. PtchHi T cells selectively homed to IRI-treated human skin xenografts to cause accelerated allograft loss, and Hh signaling was sufficient for this process to occur.\n\nDiscussion: Our studies define functional heterogeneity among a PtchHi T-cell population implicated in IRI.","prettyUrl":"wang-2023-fi","following":false,"created":"07/17/2023","featured":false,"publishedDate":"07/17/2023","urlOrId":"wang-2023-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c6516d68-8691-4864-a9b7-56cc178623cc","title":"Immunophenotypic correlates of sustained MRD negativity in patients with multiple myeloma","investigator":"Coffey, David G.","investigatorInstitution":"Sylvester Comprehensive Cancer Center, University of Miami","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"296":"coffey-2023-nc"},"prettyUrlList":["coffey-2023-nc"],"summary":"The role of the immune microenvironment in maintaining disease remission in patients with multiple myeloma (MM) is not well understood. We comprehensively profiled the immune system in patients with newly diagnosed MM receiving continuous lenalidomide maintenance therapy with the aim of uncovering correlates of long-term treatment response. Leveraging single-cell RNA sequencing and T cell receptor β sequencing of the peripheral blood and CyTOF mass cytometry of the bone marrow, we longitudinally characterized the immune landscape in 23 patients before and one year after lenalidomide exposure. We compared patients achieving sustained minimal residual disease (MRD) negativity to patients who never achieved or were unable to maintain MRD negativity. We observed that the composition of the immune microenvironment in both the blood and the marrow varied substantially according to both MRD negative status and history of autologous stem cell transplant, supporting the hypothesis that the immune microenvironment influences the depth and duration of treatment response.","prettyUrl":"coffey-2023-nc","following":false,"created":"12/06/2022","featured":false,"publishedDate":"06/30/2023","urlOrId":"coffey-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"fb2f87ff-6d7b-4b4a-afee-3c78b185e610","title":"Phototherapy restores deficient type I interferon production and enhances antitumor responses in mycosis fungoides","investigator":"Clark, Rachael","investigatorInstitution":"Brigham and Women's Hospital","publicationName":"Journal of Investigative Dermatology","researchArea":"Cancer","prettyUrls":{"295":"clark-2023-jid"},"prettyUrlList":["clark-2023-jid"],"summary":"Transcriptional profiling demonstrated markedly reduced type I IFN gene expression in untreated mycosis fungoides (MF) skin lesions compared with that in healthy skin. Type I IFN expression in MF correlated with antigen-presenting cell-associated IRF5 before psoralen plus UVA therapy and epithelial ULBP2 after therapy, suggesting an enhancement of epithelial type I IFN. Immunostains confirmed reduced baseline type I IFN production in MF and increased levels after psoralen plus UVA treatment in responding patients. Effective tumor clearance was associated with increased type I IFN expression, enhanced recruitment of CD8+ T cells into skin lesions, and expression of genes associated with antigen-specific T-cell activation. IFNk, a keratinocyte-derived inducer of type I IFNs, was increased by psoralen plus UVA therapy and expression correlated with upregulation of other type I IFNs. In vitro, deletion of keratinocyte IFNk decreased baseline and UVA-induced expression of type I IFN and IFN response genes. In summary, we find a baseline deficit in type I IFN production in MF that is restored by psoralen plus UVA therapy and correlates with enhanced antitumor responses. This may explain why MF generally develops in sun-protected skin and suggests that drugs that increase epithelial type I IFNs, including topical MEK and EGFR inhibitors, may be effective therapies for MF.","prettyUrl":"clark-2023-jid","following":false,"created":"06/15/2023","featured":false,"publishedDate":"06/22/2023","urlOrId":"clark-2023-jid","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"24e260ad-9956-4062-a9a1-7b28b803dc54","title":"The overlap of blood and skin T-cell clones in early-stage mycosis fungoides","investigator":"Nikbakht, Neda","investigatorInstitution":"Thomas Jefferson University, Department of Dermatology and Cutaneous Biology","publicationName":"Blood Advances","researchArea":"Cancer","prettyUrls":{"294":"joffe-2023-ba"},"prettyUrlList":["joffe-2023-ba"],"summary":"Mycosis fungoides (MF) is the most common subtype of cutaneous T-cell lymphoma that initially starts in the skin but can progress to involve blood with significant mortality in late stages. The presence of a dominant T-cell clone in the blood of early-stage MF appears to be associated with a poor prognosis. However, the identity of dominant blood T-cell clones and the degree of overlap between T-cell repertoires of blood and skin in MF have not been fully interrogated. Here, we evaluated the relationship of blood and skin T-cell repertoires and their relevance to prognosis in MF. We used high-throughput sequencing to interrogate the T-cell receptor (TCR) complementarity determining region 3 sequences in blood and skin of MF patients at the Jefferson Cutaneous Lymphoma Clinic (Adaptive Biotechnologies). ImmunoSEQ Analyzer provided T-cell repertoire overlap metric (Morisita’s index) and diversity measure (Simpson’s clonality score). Time to systemic treatment (TTST) was calculated as the time from initial diagnosis to initiation of first systemic therapy. \n60 MF patients with no blood involvement were enrolled. 28% had a dominant clone in blood; of these, 18% were identical to dominant skin clones and 82% were distinct from the dominant clones identified in skin. We found that MF patients with discordant dominant TCR clones in blood and skin had a longer TTST when compared to the identical cohort (P=0.0057). Furthermore, patients with discordant dominant clones had a lower degree of T-cell repertoire overlap between blood and skin (P<0.0001) and higher blood T-cell repertoire diversity scores (P=0.013) when compared to the identical group. Our data shows that MF patients with dominant clones in blood segregate into two cohorts with distinct prognoses based on the identity of their peripheral T-cell clones.","prettyUrl":"joffe-2023-ba","following":false,"created":"06/07/2023","featured":false,"publishedDate":"06/20/2023","urlOrId":"joffe-2023-ba","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d6a06643-4f97-4de7-aa3d-d1f3f0d0b6d1","title":"Mesothelin-targeting T cell receptor fusion construct cell therapy in refractory solid tumors: phase 1/2 trial interim results","investigator":"Hassan, Raffit","investigatorInstitution":"Thoracic and GI Malignancies Branch, NCI","publicationName":"Nature Medicine","researchArea":"Cancer","prettyUrls":{"293":"hassan-2023-nm"},"prettyUrlList":["hassan-2023-nm"],"summary":"The T cell receptor fusion construct (TRuC) gavocabtagene autoleucel (gavo-cel) consists of single-domain anti-mesothelin antibody that integrates into the endogenous T cell receptor (TCR) and engages the signaling capacity of the entire TCR upon mesothelin binding. Here we describe phase 1 results from an ongoing phase1/2 trial of gavo-cel in patients with treatment-refractory mesothelin-expressing solid tumors. The primary objectives were to evaluate safety and determine the recommended phase 2 dose (RP2D). Secondary objectives included efficacy. Thirty-two patients received gavo-cel at increasing doses either as a single agent (n = 3) or after lymphodepletion (LD, n = 29). Dose-limiting toxicities of grade 3 pneumonitis and grade 5 bronchioalveolar hemorrhage were noted. The RP2D was determined as 1 × 108 cells per m2 after LD. Grade 3 or higher pneumonitis was seen in 16% of all patients and in none at the RP2D; grade 3 or higher cytokine release syndrome occurred in 25% of all patients and in 15% at the RP2D. In 30 evaluable patients, the overall response rate and disease control rate were 20% (13% confirmed) and 77%, respectively, and the 6-month overall survival rate was 70%. Gavo-cel warrants further study in patients with mesothelin-expressing cancers given its encouraging anti-tumor activity, but it may have a narrow therapeutic window. ClinicalTrials.gov identifier: NCT03907852 .","prettyUrl":"hassan-2023-nm","following":false,"created":"06/01/2023","featured":false,"publishedDate":"06/01/2023","urlOrId":"hassan-2023-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b8360822-76fc-47a3-b453-6f7ed056c0f5","title":"Differentiation of exhausted CD8 T cells after termination of chronic antigen stimulation stops short of achieving functional T cell memory","investigator":"Lauer, Georg M.","investigatorInstitution":"Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School","publicationName":"Nature Immunology","researchArea":"Other","prettyUrls":{"292":"tonnerre-2021-ni"},"prettyUrlList":["tonnerre-2021-ni"],"summary":"T cell exhaustion is associated with failure to clear chronic infections and malignant cells. Defining the molecular mechanisms of T cell exhaustion and reinvigoration is essential to improving immunotherapeutic modalities. Analysis of antigen-specific CD8+ T cells before and after antigen removal in human hepatitis c virus (HCV) infection confirmed pervasive phenotypic, functional, and transcriptional differences between exhausted and memory CD8+ T cells. After viral cure, we observed broad phenotypic and transcriptional changes in clonally stable exhausted T-cell populations suggesting differentiation towards a memory-like profile. However, functionally, the cells showed little improvement and critical transcriptional regulators remained in the exhaustion state. Notably, T cells from chronic HCV infection that were exposed to antigen for shorter periods of time because of viral escape mutations were functionally and transcriptionally more similar to memory T cells from spontaneously resolved acute HCV infection. Thus, duration of T cell stimulation impacts the ability to recover from exhaustion, as antigen removal after long-term T cell exhaustion is insufficient for the development of key T cell memory characteristics.","prettyUrl":"tonnerre-2021-ni","following":false,"created":"06/04/2021","featured":false,"publishedDate":"05/24/2023","urlOrId":"tonnerre-2021-ni","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"e69bc69b-98b5-4585-a220-afd95f731170","title":"Perturbations of the T-cell immune repertoire in kidney transplant rejection","investigator":"Sarwal, Minnie M","investigatorInstitution":"Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco","publicationName":"Frontiers in Immunology","researchArea":"Organ transplant","prettyUrls":{"290":"sigdel-2022-fi"},"prettyUrlList":["sigdel-2022-fi"],"summary":"In this cross-sectional and longitudinal analysis of mapping the T-cell repertoire in kidney transplant recipients, we have investigated and validated T-cell clonality, immune repertoire chronology at rejection, and contemporaneous allograft biopsy quantitative tissue injury, to better understand the pathobiology of acute T-cell fraction, T-cell repertoire and antibody-mediated kidney transplant rejection. To follow the dynamic evolution of T-cell repertoire changes before and after engraftment and during biopsy-confirmed acute rejection, we sequenced 323 peripheral blood samples from 200 unique kidney transplant recipients, with (n=100) and without (n=100) biopsy-confirmed acute rejection. We report that patients who develop acute allograft rejection, have lower (p=0.01) T-cell fraction even before transplantation, followed by its rise after transplantation and at the time of acute rejection accompanied by high TCR repertoire turnover (p=0.004). Acute rejection episodes occurring after the first 6 months post-transplantation, and those with a component of antibody-mediated rejection, had the highest turnover; p=0.0016) of their T-cell repertoire. In conclusion, we validated that detecting repertoire changes in kidney transplantation correlates with post-transplant rejection episodes suggesting that T-cell receptor sequencing may provide recipient pre-transplant and post-transplant predictors of rejection risk.","prettyUrl":"sigdel-2022-fi","following":false,"created":"04/24/2023","featured":false,"publishedDate":"05/23/2023","urlOrId":"sigdel-2022-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"e05475e7-2695-4b91-b89e-64de88cdc6ab","title":"Inherited ARPC5 mutations cause an actinopathy impairing cell motility and disrupting cytokine signaling","investigator":"Rosenzweig, Sergio D.","investigatorInstitution":"Immunology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health","publicationName":"Nature Communications","researchArea":"Immunocompetence","prettyUrls":{"291":"nunes-santos-2023-nc"},"prettyUrlList":["nunes-santos-2023-nc"],"summary":"We describe the first cases of germline biallelic null mutations in ARPC5, part of the Arp2/3 actin nucleator complex, in two unrelated patients presenting with recurrent and severe infections, early-onset autoimmunity, inflammation, and dysmorphisms. This defect compromises multiple cell lineages and functions, and when protein expression is reestablished in-vitro, the Arp2/3 complex conformation and functions are rescued. As part of the pathophysiological evaluation, we also show that interleukin (IL)-6 signaling is distinctively impacted in this syndrome. Disruption of IL-6 classical but not trans-signaling highlights their differential roles in the disease and offers perspectives for therapeutic molecular targets.","prettyUrl":"nunes-santos-2023-nc","following":false,"created":"11/10/2022","featured":false,"publishedDate":"05/22/2023","urlOrId":"nunes-santos-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b11ccf4a-cca7-452b-8345-b0b23c565855","title":"The prospect of universal coronavirus immunity: characterization of reciprocal and non-reciprocal T cell responses against SARS-CoV2 and common human coronaviruses","investigator":"Soni, Mithil","investigatorInstitution":"Columbia Center for Translational Immunology, Department of Medicine, Columbia University","publicationName":"Frontiers in Immunology","researchArea":"Infectious Disease","prettyUrls":{"289":"mithil-2023-fi"},"prettyUrlList":["mithil-2023-fi"],"summary":"T cell immunity plays a central role in clinical outcomes of Coronavirus Infectious Disease 2019 (COVID-19). Therefore, T cell-focused vaccination or cellular immunotherapy might provide enhanced protection for immunocompromised patients. Pre-existing T cell memory recognizing SARS-CoV2 antigens antedating COVID-19 infection or vaccination, may have developed as an imprint of prior infections with endemic non-SARS human coronaviruses (hCoVs) OC43, HKU1, 229E, NL63, pathogens of “common cold”. In turn, SARS-CoV2-primed T cells may recognize emerging variants or other hCoV viruses and modulate the course of subsequent hCoV infections. Cross-immunity between hCoVs and SARS-CoV2 has not been well characterized. Here, we systematically investigated T cell responses against the immunodominant SARS-CoV2 spike, nucleocapsid and membrane proteins and corresponding antigens from α- and β-hCoVs among vaccinated, convalescent, and unexposed subjects. Broad T cell immunity against all tested SARS-CoV2 antigens emerged in COVID-19 survivors. In convalescent and in vaccinated individuals, SARS-CoV2 spike-specific T cells reliably recognized most SARS-CoV2 variants, however cross-reactivity against the omicron variant was reduced by approximately 50%. Responses against spike, nucleocapsid and membrane antigens from endemic hCoVs were more extensive in COVID-19 survivors than in unexposed subjects and displayed cross-reactivity between α- and β-hCoVs. In some, non-SARS hCoV-specific T cells demonstrated a prominent non-reciprocal cross-reactivity with SARS-CoV2 antigens, whereas a distinct anti-SARS-CoV2 immunological repertoire emerged post-COVID-19, with relatively limited cross-recognition of non-SARS hCoVs. Based on this cross-reactivity pattern, we established a strategy for in-vitro expansion of universal anti-hCoV T cells for adoptive immunotherapy. Overall, these results have implications for the future design of universal vaccines and cell-based immune therapies against SARS- and non-SARS-CoVs.","prettyUrl":"mithil-2023-fi","following":false,"created":"04/21/2023","featured":false,"publishedDate":"04/21/2023","urlOrId":"mithil-2023-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"8129cdf8-cd65-4136-8a4c-32f145f10a27","title":"Reduced skin T-cell receptor diversity in large cell transformed mycosis fungoides","investigator":"Nikbakht, Neda","investigatorInstitution":"Department of Dermatology and Cutaneous Biology, Thomas Jefferson University","publicationName":"Journal of Investigative Dermatology","researchArea":"Dermatology","prettyUrls":{"288":"gleason-2023-jid"},"prettyUrlList":["gleason-2023-jid"],"summary":"We assess changes in the T-cell repertoire diversity in an aggressive subtype of cutaneous T-cell lymphoma. Mycosis fungoides (MF) is a type of T-cell lymphoma that originates in the skin and can progress to extracutaneous sites in late stages. Large cell transformed mycosis fungoides (LCT-MF) is an aggressive subtype of MF associated with a higher likelihood of malignancy progression and mortality. Given that T-cell repertoire diversity and skin tumor cell frequency (TCF) may influence prognosis, we sought to investigate these metrics in LCT-MF. We sequenced the complementarity determining regions of T-cell receptor genes in skin biopsies and blood samples of aged-matched healthy subjects, non-leukemic MF and LCT-MF. Overall, our results demonstrate that while lesional skin of LCT-MF differs from MF in clonal diversity in both late and early-stage, its malignant T-cell frequency differs only in early-stage. Our data can provide insight into the tissue specific origin of early T-cell transformation events in MF.","prettyUrl":"gleason-2023-jid","following":false,"created":"04/10/2023","featured":false,"publishedDate":"04/19/2023","urlOrId":"gleason-2023-jid","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"f1e157b6-527f-47a5-aae9-6b48aa7978b5","title":"Neoadjuvant enoblituzumab in localized prostate cancer: a single arm, phase 2 trial","investigator":"Shenderov, Eugene","investigatorInstitution":"Department of Oncology, Johns Hopkins School of Medicine","publicationName":"Nature Medicine","researchArea":"Cancer","prettyUrls":{"287":"shenderov-2023-nm"},"prettyUrlList":["shenderov-2023-nm"],"summary":"B7 homolog 3 (B7-H3; CD276), a tumor-associated antigen and possible immune checkpoint, is highly expressed in prostate cancer (PCa) and is associated with early recurrence and metastasis. Enoblituzumab is a humanized, Fc-engineered, B7-H3-targeting antibody that mediates antibody-dependent cellular cytotoxicity. In this phase 2, biomarker-rich neoadjuvant trial, 32 biological males with operable intermediate to high-risk localized PCa were enrolled to evaluate the safety, anti-tumor activity and immunogenicity of enoblituzumab when given before prostatectomy. The coprimary outcomes were safety and undetectable prostate-specific antigen (PSA) level (PSA0) 1 year postprostatectomy, and the aim was to obtain an estimate of PSA0 with reasonable precision. The primary safety endpoint was met with no notable unexpected surgical or medical complications, or surgical delay. Overall, 12% of patients experienced grade 3 adverse events and no grade 4 events occurred. The coprimary endpoint of the PSA0 rate 1 year postprostatectomy was 66% (95% confidence interval 47-81%). The use of B7-H3-targeted immunotherapy in PCa is feasible and generally safe and preliminary data suggest potential clinical activity. The present study validates B7-H3 as a rational target for therapy development in PCa with larger studies planned. The ClinicalTrials.gov identifier is NCT02923180.","prettyUrl":"shenderov-2023-nm","following":false,"created":"02/27/2023","featured":false,"publishedDate":"04/14/2023","urlOrId":"shenderov-2023-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"891e78f0-95af-4e62-8910-ade6c90520a2","title":"Targeting advanced prostate cancer with STEAP1 chimeric antigen receptor T cell and tumor-localized IL-12 immunotherapy","investigator":"Lee, John K","investigatorInstitution":"Fred Hutchinson Cancer Center and University of Washington","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"286":"bhatia-2023-nc"},"prettyUrlList":["bhatia-2023-nc"],"summary":"Six transmembrane epithelial antigen of the prostate 1 (STEAP1) is a compelling cell surface antigen for therapeutic targeting in prostate cancer. We report broad expression of STEAP1 relative to prostate-specific membrane antigen (PSMA) in lethal metastatic prostate cancers and the development of a STEAP1-directed chimeric antigen receptor (CAR) T cell therapy. STEAP1 CAR T cells demonstrate reactivity in low antigen density, antitumor activity across metastatic prostate cancer models, and safety in a human STEAP1 knock-in mouse model. STEAP1 antigen escape is a recurrent mechanism of treatment resistance and is associated with diminished tumor antigen processing and presentation. The application of tumor-localized interleukin-12 (IL-12) therapy in the form of a collagen binding domain (CBD)-IL-12 fusion protein combined with STEAP1 CAR T cell therapy enhances antitumor efficacy by remodeling the immunologically cold tumor microenvironment of prostate cancer and combating STEAP1 antigen escape through the engagement of host immunity and epitope spreading.","prettyUrl":"bhatia-2023-nc","following":false,"created":"03/30/2023","featured":false,"publishedDate":"03/30/2023","urlOrId":"bhatia-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"ad7a2d37-a0bc-4d88-813e-6dd7d762a65b","title":"T cell receptor repertoire sequencing reveals chemotherapy-driven clonal expansion in colorectal liver metastases","investigator":"Flatmark, Kjersti","investigatorInstitution":"Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital","publicationName":"GigaScience","researchArea":"Cancer","prettyUrls":{"285":"hoye-2023-gs"},"prettyUrlList":["hoye-2023-gs"],"summary":"Background: Colorectal liver metastasis (CLM) is a leading cause of colorectal cancer mortality, and the response to immune checkpoint inhibition (ICI) in microsatellite stable CRC has been disappointing. Administration of cytotoxic chemotherapy may cause increased density of tumour infiltrating T cells, which has been associated with improved response to ICI. This study aimed to quantify and characterize T cell infiltration in CLM using T cell receptor (TCR) repertoire sequencing.\n\nMethods: Eighty-five resected CLM from patients included in the Oslo CoMet study were subjected to TCR repertoire sequencing. Thirty-five and 15 patients had received neoadjuvant chemotherapy (NACT) within a short or long interval, respectively, prior to resection, while 35 patients had not been exposed to NACT. T cell fractions were calculated, repertoire clonality was analysed based on Hill evenness curves, and TCR sequential convergence was assessed using network analysis.\n\nFindings: Increased T cell fractions (10·6% vs 6·3%) were detected in CLM exposed to NACT within a short interval prior to resection, while modestly increased clonality was observed in NACT exposed tumours independently of the timing of NACT administration and surgery. While private clones made up >90% of detected clones, network connectivity analysis revealed that public clones contributed the majority of TCR sequence convergence.\n\nInterpretation: TCR repertoire sequencing can be used to quantify T cell infiltration and clonality in clinical samples. This study provides evidence to support chemotherapy-driven T cell clonal expansion in CLM in a clinical context.","prettyUrl":"hoye-2023-gs","following":false,"created":"09/22/2022","featured":false,"publishedDate":"02/27/2023","urlOrId":"hoye-2023-gs","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"88f526f9-93e2-459a-aaab-e7572da91e74","title":"Microbial peptides activate tumor-infiltrating lymphocytes in glioblastoma","investigator":"Naghavian,Reza","investigatorInstitution":"Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich","publicationName":"Nature","researchArea":"Cancer Immunotherapy","prettyUrls":{"284":"naghavian-2023-n"},"prettyUrlList":["naghavian-2023-n"],"summary":"Microbial organisms play key roles in numerous physiological processes in the human body and have recently been shown to modify the response to cancer radiotherapy and immune checkpoint inhibitors. Here, we aim to address the role of microbial organisms and their potential role in immune reactivity against glioblastoma. We demonstrate that HLA molecules of both glioblastoma tissues and tumor cell lines present bacteria-specific peptides. This finding prompted us to examine whether tumor-infiltrating lymphocytes (TILs) recognize tumor-derived bacterial peptides. Bacterial peptides eluted from HLA-II molecules are recognized by TILs, albeit very weakly. Using an unbiased antigen discovery approach to probe the specificity of a TIL CD4+ T cell clone, we show that it recognizes a broad spectrum of peptides from pathogenic bacteria, commensal gut microbiota and also glioblastoma-related tumor antigens. These peptides were also strongly stimulatory for bulk TILs and peripheral blood memory cells, which then respond to tumor-derived target peptides. Our data hint at how bacterial pathogens and bacterial gut microbiota can be involved in specific immune recognition of tumor antigens. The unbiased identification of microbial target antigens for TILs holds promise for future personalized tumor vaccination approaches.","prettyUrl":"naghavian-2023-n","following":false,"created":"02/14/2023","featured":false,"publishedDate":"02/27/2023","urlOrId":"naghavian-2023-n","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1405901d-9e88-4c21-9f4a-07f530b2aafa","title":"Clonally expanded HIV-1 proviruses with 5’-Leader defects can give rise to nonsuppressible residual viremia","investigator":"Simonetti, Francesco R.","investigatorInstitution":"Department of Medicine, Johns Hopkins University School of Medicine","publicationName":"Journal of Clinical Investigation","researchArea":"Infectious Disease","prettyUrls":{"283":"white-2023-jci"},"prettyUrlList":["white-2023-jci"],"summary":"Background: Antiretroviral therapy (ART) halts HIV-1 replication, decreasing viremia to below the detection limit of clinical assays. However, some individuals experience persistent nonsuppressible viremia (NSV) originating from CD4+ T cell clones carrying infectious proviruses. Defective proviruses represent over 90% of all proviruses persisting during ART and can express viral genes, but whether they can cause NSV and complicate ART management is unknown.\n\nMethods: We carried an in-depth characterization of proviruses causing NSV in 4 study participants with optimal adherence and no drug resistance. We investigated the impact of the observed defects on 5'-Leader RNA properties, virus infectivity, and gene expression. Integration-site specific assays were used to track these proviruses over time and among cell subsets.\n\nResults: Clones carrying proviruses with 5'-Leader defects can cause persistent NSV up to ~103 copies/mL. These proviruses had small, often identical deletions or point mutations involving the major splicing donor site (MSD) and showed partially reduced RNA dimerization and nucleocapsid binding. Nevertheless, they were inducible and produced non-infectious virions containing viral RNA but lacking Envelope.\n\nConclusion: These findings show that proviruses with 5'-Leader defects in CD4+ T cell clones can give rise to NSV, affecting clinical care. Sequencing of the 5'-Leader can help understanding failure to completely suppress viremia.","prettyUrl":"white-2023-jci","following":false,"created":"12/19/2022","featured":false,"publishedDate":"02/15/2023","urlOrId":"white-2023-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"9a3204c8-3241-4dce-bb6a-7af4cc6be9ef","title":"Combination IFNβ and membrane-stable CD40L maximize tumor dendritic cell activation and lymph node trafficking to elicit systemic T-cell immunity","investigator":"Zheng, Hong","investigatorInstitution":"Moffitt Cancer Center and Research Institute","publicationName":"Cancer immunology research","researchArea":"Other","prettyUrls":{"282":"zheng-2023-cir"},"prettyUrlList":["zheng-2023-cir"],"summary":"Oncolytic viral therapies induce direct killing of tumor cells and activation of conventional dendritic cells (cDCs); however, cDC activation has not been optimized with current therapies. We evaluated adenoviral delivery of engineered membrane-stable CD40L (MEM40) and IFNβ to locally activate cDCs in mouse tumor models. Combined tumor MEM40 and IFNβ expression induced the highest cDC activation coupled with increased lymph node migration, increased systemic antitumor CD8+ T-cell responses, and regression of established tumors in a cDC1-dependent manner. MEM40+IFNβ combined with checkpoint inhibitors led to effective control of distant tumors and lung metastases. An oncolytic adenovirus (MEM-288) expressing MEM40+IFNβ in phase 1 clinical testing induced cancer cell loss concomitant with enhanced T-cell infiltration and increased systemic presence of tumor T-cell clonotypes in NSCLC patients. This approach to simultaneously target two major DC-activating pathways has potential to significantly impact the solid tumor immunotherapy landscape.","prettyUrl":"zheng-2023-cir","following":false,"created":"01/20/2023","featured":false,"publishedDate":"02/14/2023","urlOrId":"zheng-2023-cir","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"218392a3-618b-4fbf-8785-3509684f8613","title":"A phase Ib trial evaluating the safety, efficacy, and immunologic effects of pembrolizumab plus paclitaxel or flat-dose capecitabine in 1st/2nd line metastatic triple-negative breast cancer","investigator":"Page, David B.","investigatorInstitution":"Earle A. Chiles Research Institute, Providence Cancer Institute","publicationName":"NPJ Breast Cancer","researchArea":"Cancer","prettyUrls":{"281":"page-2023-npj"},"prettyUrlList":["page-2023-npj"],"summary":"Chemoimmunotherapy with anti-programmed cell death 1/ligand 1 and cytotoxic chemotherapy is a promising therapeutic modality for women with triple-negative breast cancer, but questions remain regarding optimal chemotherapy backbone and biomarkers for patient selection. We report final outcomes from a phase Ib trial evaluating pembrolizumab (200mg IV every 3 weeks) with either weekly paclitaxel (80mg/m2 weekly) or flat-dose capecitabine (2000mg orally twice daily for 7 days of every 14-day cycle) in the 1st/2nd line setting. The primary endpoint is safety (receipt of 2 cycles without grade III/IV toxicities requiring discontinuation or ≥21-day delays). The secondary endpoint is efficacy (week 12 objective response). Exploratory aims are to characterize immunologic effects of treatment over time, and to evaluate novel biomarkers. The trial demonstrates that both regimens meet the pre-specified safety endpoint (paclitaxel: 87%; capecitabine: 100%). Objective response rate is 29% for pembrolizumab/paclitaxel (n=4/13, 95% CI: 10-61%) and 43% for pembrolizumab/capecitabine (n=6/14, 95% CI: 18-71%). Partial responses are observed in two subjects with chemo-refractory metaplastic carcinoma (both in capecitabine arm). Both regimens are associated with significant peripheral leukocyte contraction over time. Response is associated with clinical PD-L1 score, non-receipt of prior chemotherapy, and the H&E stromal tumor infiltrating lymphocyte score, but also by a novel 27 gene IO score and spatial biomarkers (lymphocyte spatial skewness). In conclusion, pembrolizumab with paclitaxel or capecitabine is safe and clinically active. Both regimens are lymphodepleting, highlighting the competing immunostimulatory versus lymphotoxic effects of cytotoxic chemotherapy. Further exploration of the IO score and spatial TIL biomarkers is warranted.","prettyUrl":"page-2023-npj","following":false,"created":"12/21/2022","featured":false,"publishedDate":"02/10/2023","urlOrId":"page-2023-npj","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"2457f729-fa61-429c-a97b-6b0e1aed9130","title":"Systematic lineage tracing reveals clonal progenitors and long-term persistence of tumor-specific T cells during immune checkpoint blockade.","investigator":"Andrew Chow","investigatorInstitution":"Memorial Sloan Kettering Cancer Center","publicationName":"Cancer Cell","researchArea":"Cancer Immunotherapy","prettyUrls":{"280":"pai-2023-cc"},"prettyUrlList":["pai-2023-cc"],"summary":"Paired single-cell RNA and T cell receptor sequencing (scRNA/TCR-seq) has allowed\nfor enhanced resolution of clonal T cell dynamics in cancer. Here, we report\nscRNA/TCR-seq analysis of 187,650 T cells from 31 tissue regions, including tumor,\nadjacent normal tissues, and lymph nodes (LN), from patients who underwent\nresections for persistent or progressing lung cancers after immune checkpoint\nblockade (ICB). We found marked regional heterogeneity in CD8+ and CD4+ T cell\nphenotypes that was associated with heterogeneity in the presence of viable tumor\ncells. Regions with viable cancer cells were enriched for exhausted CD8+ T cells,\nregulatory CD4+ T cells (Treg), and follicular helper CD4+ T cells (TFH). Tracking\nindividual T cell clonotypes across tumor regions and tissues, combined with\nneoantigen specificity assays, revealed that TFH and tumor-specific exhausted CD8+\nT cells could be clonally linked to TCF7+ SELL+ progenitors in tumor draining LNs, and\nprogressive exhaustion trajectories of CD8+ T, Treg, and TFH cells with proximity to\nthe tumor microenvironment. Finally, longitudinal tracking of tumor-specific CD8+ and\nCD4+ T cell clones revealed persistence in the peripheral blood for years after ICB\ntherapy; however, exhausted CD8+, Treg, and TFH cells had lower persistence relative\nto other subsets. Strikingly, the presence of clonally linked progenitors in the LN\nconferred greater longitudinal persistence. Altogether, this comprehensive\nscRNA/TCR-seq dataset with regional, clonal, and longitudinal resolution provides\nfundamental insights into the tissue distribution, differentiation trajectories, and\npersistence of the tumor-specific T cells that underlie clinical responses to ICB.","prettyUrl":"pai-2023-cc","following":false,"created":"02/05/2023","featured":false,"publishedDate":"02/09/2023","urlOrId":"pai-2023-cc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"bed85d2c-c678-4ec7-b243-8e55588a7ca5","title":"Single-cell characterization of anti-LAG3+anti-PD1 treatment in melanoma patients","investigator":"Huuhtanen, Jani","investigatorInstitution":"Hematology Research Unit Helsinki, University of Helsinki","publicationName":"Journal of Clinical Investigation","researchArea":"Cancer","prettyUrls":{"279":"huuhtanen-2023-jci"},"prettyUrlList":["huuhtanen-2023-jci"],"summary":"Background: Relatlimab+nivolumab (anti-LAG3+anti-PD1) has been approved by FDA as a 1st-line therapy in stage III/IV melanoma, but its detailed effect on the immune system is unknown. \n\nMethods: We evaluated blood samples from 40 immunotherapy-naïve or prior immunotherapy-refractory patients with metastatic melanoma treated with anti-LAG3+anti-PD1 in a phase I trial (NCT01968109) using single-cell RNA and T cell receptor (TCR) sequencing (scRNA+TCRαβ-seq) combined with other multiomics profiling. \n\nResults: The highest LAG3 expression was noted in NK cells, regulatory T cells (Tregs), and CD8+ T cells, and these cell populations underwent the most significant changes during the treatment. Adaptive NK cells were enriched in responders and underwent profound transcriptomic changes during the therapy resulting in an active phenotype. LAG3+ Tregs expanded but based on the transcriptome profile became metabolically silent during the treatment. Lastly, higher baseline TCR clonality was observed in responding patients, and their expanding CD8+ T cell clones gained more cytotoxic and NK-like phenotype. \n\nConclusion: Anti-LAG3+anti-PD1 therapy has profound effects on NK cells and Tregs in addition to CD8+ T cells.","prettyUrl":"huuhtanen-2023-jci","following":false,"created":"02/07/2023","featured":false,"publishedDate":"02/08/2023","urlOrId":"huuhtanen-2023-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7f1ac13d-292c-43f6-a12e-4534a12900e1","title":"Impaired SARS-CoV-2 variant neutralization and CD8+ T cell responses following 3 doses of mRNA vaccines in myeloma: correlation with breakthrough infections","investigator":"Dhodapkar, Madhav V.","investigatorInstitution":"Department of Hematology/Medical Oncology, Emory University","publicationName":"Blood Cancer Discovery","researchArea":"Infectious Disease","prettyUrls":{"278":"azeem-2023-bcd"},"prettyUrlList":["azeem-2023-bcd"],"summary":"Patients with multiple myeloma (MM) mount suboptimal neutralizing antibodies (nAb) following 2 doses of SARS-CoV-2 mRNA vaccines. Currently, circulating SARS-CoV-2 variants of concern (VOC) carry the risk of breakthrough infections. We evaluated immune recognition of current VOC including BA.1, BA.2, and BA.5 in 331 racially representative patients with MM following 2 or 3 doses of mRNA vaccines. The third dose increased nAbs against WA1 in 82%, but against BA variants in only 33% to 44% of patients. Vaccine-induced nAbs correlated with receptor-binding domain (RBD)–specific class-switched memory B cells. Vaccine-induced spike-specific T cells were detected in patients without seroconversion and cross-recognized variant-specific peptides but were predominantly CD4+ T cells. Detailed clinical/immunophenotypic analysis identified features correlating with nAb/B/T-cell responses. Patients who developed breakthrough infections following 3 vaccine doses had lower live-virus nAbs, including against VOC. Patients with MM remain susceptible to SARS-CoV-2 variants following 3 vaccine doses and should be prioritized for emerging approaches to elicit variant-nAb and CD8+ T cells.","prettyUrl":"azeem-2023-bcd","following":false,"created":"02/01/2023","featured":false,"publishedDate":"02/01/2023","urlOrId":"azeem-2023-bcd","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a72d3b15-ce5a-483c-a15c-5f953a74680b","title":"Immunosequencing and profiling of T cells at the maternal-fetal interface in preterm labor and birth","investigator":"Gomez-Lopez, Nardhy","investigatorInstitution":"Department of Obstetrics and Gynecology, Wayne State University School of Medicine","publicationName":"Journal of Immunology","researchArea":"Immunocompetence","prettyUrls":{"277":"miller-2023-ji"},"prettyUrlList":["miller-2023-ji"],"summary":"T cells are implicated in the pathophysiology of preterm labor and birth, the leading cause of neonatal morbidity and mortality worldwide. Specifically, maternal decidual T cells infiltrate the chorioamniotic membranes in chronic chorioamnionitis (CCA), a placental lesion considered to reflect maternal anti-fetal rejection, leading to preterm labor and birth. However, the phenotype and TCR repertoire of decidual T cells in women with preterm labor and CCA have not been investigated. In this study, we used phenotyping, TCR sequencing, and functional assays to elucidate the molecular characteristics and Ag specificity of T cells infiltrating the chorioamniotic membranes in women with CCA who underwent term or preterm labor. Phenotyping indicated distinct enrichment of human decidual effector memory T cell subsets in cases of preterm labor with CCA without altered regulatory T cell proportions. TCR sequencing revealed that the T cell repertoire of CCA is characterized by increased TCR richness and decreased clonal expansion in women with preterm labor. We identified 15 clones associated with CCA and compared these against established TCR databases, reporting that infiltrating T cells may possess specificity for maternal and fetal Ags, but not common viral Ags. Functional assays demonstrated that choriodecidual T cells can respond to maternal and fetal Ags. Collectively, our findings provide, to our knowledge, novel insight into the complex processes underlying chronic placental inflammation and further support a role for effector T cells in the mechanisms of disease for preterm labor and birth. Moreover, this work further strengthens the contribution of adaptive immunity to the syndromic nature of preterm labor and birth.","prettyUrl":"miller-2023-ji","following":false,"created":"01/30/2023","featured":false,"publishedDate":"02/01/2023","urlOrId":"miller-2023-ji","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a1b2d1a0-e3d1-4e6e-9669-e004ed04f4b8","title":"Clonal dynamics of alloreactive T cells in kidney allograft rejection after anti-PD-1 therapy","investigator":"Murakami, Naoka","investigatorInstitution":"Brigham and Women's Hospital, Division of Renal Medicine","publicationName":"Nature Communications","researchArea":"Organ transplant","prettyUrls":{"276":"dunlap-2023-nc"},"prettyUrlList":["dunlap-2023-nc"],"summary":"Kidney transplant recipients are at particular risk for developing tumors, many of which are now routinely treated with immune checkpoint inhibitors (ICIs); however, ICI therapy can precipitate transplant rejection. We utilized TCR sequencing to identify and track alloreactive T cells in a patient with melanoma who experienced kidney transplant rejection following ICI therapy. ICI therapy was associated with a sharp increase in circulating alloreactive CD8+ T cell clones, many of which were also detected in the rejected kidney but not at tumor sites. Longitudinal and cross-tissue TCR analyses revealed unintended expansion of alloreactive CD8+ T cells induced by ICI therapy for cancer, coinciding with ICI-associated organ rejection.","prettyUrl":"dunlap-2023-nc","following":false,"created":"10/29/2022","featured":false,"publishedDate":"01/30/2023","urlOrId":"dunlap-2023-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"885ef16f-5c7a-437a-a6cc-01872b6c8c58","title":"Host-specific differences in top-expanded TCR clonotypes correlate with divergent outcomes of anti-PD-L1 treatment in responders versus non-responders","investigator":"Wang, Jing H","investigatorInstitution":"Department of Medicine, University of Pittsburgh","publicationName":"Frontiers in Immunology","researchArea":"Cancer Immunotherapy","prettyUrls":{"275":"john-2023-fi"},"prettyUrlList":["john-2023-fi"],"summary":"Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment; however, the responses to ICI treatment are highly variable in different individuals and the underlying mechanisms remain poorly understood. Here, we employed a mouse squamous cell carcinoma (SCC) model where tumor-bearing recipients diverged into responders (R) versus non-responders (NR) upon anti-PD-L1 treatment. We performed in-depth TCR sequencing with immunoSEQ platform to delineate the differences in CD8 tumor-infiltrating lymphocytes (TILs). We found that R and NR CD8 TILs both exhibited evidence of clonal expansion, suggesting activation regardless of response status. We detected no differences in clonal expansion or clonal diversity indexes between R vs. NR. However, the top expanded (>1%) TCR clonotypes appeared to be mutually exclusive between R and NR CD8 TILs, showing a preferential expansion of distinct TCR clonotypes in response to the same SCC tumor in R vs. NR. Notably, the mutual exclusivity of TCR clonotypes in R vs. NR was only observed when top TCR clonotypes were counted, because such top-expanded clonotypes are present in the opposite outcome group at a much lower frequency. Many TCR sequences were detected in only one recipient at a high frequency, implicating highly individualized anti-tumor immune responses. We conclude that differences in the clonal frequency of top TCR clonotypes between R and NR CD8 TILs may be one of the factors underlying differential anti-PD-L1 responses. This notion may offer a novel explanation for variable ICI responses in different individuals, which may substantially impact the development of new strategies for personalized cancer immunotherapy.","prettyUrl":"john-2023-fi","following":false,"created":"01/30/2023","featured":false,"publishedDate":"01/30/2023","urlOrId":"john-2023-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"91a8f980-42f0-4385-b8eb-ec5efddf54cf","title":"Immune environment and antigen specificity of the T cell receptor repertoire of malignant ascites in ovarian cancer","investigator":"Klopp, Ann H.","investigatorInstitution":"Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center","publicationName":"PLOS ONE","researchArea":"Cancer","prettyUrls":{"274":"court-2023-po"},"prettyUrlList":["court-2023-po"],"summary":"We evaluated the association of disease outcome with T cell immune-related characteristics and T cell receptor (TCR) repertoire in malignant ascites from patients with high-grade epithelial ovarian cancer. Ascitic fluid samples were collected from 47 high-grade epithelial ovarian cancer patients and analyzed using flow cytometry and TCR sequencing to characterize the complementarity determining region 3 TCR β-chain. TCR functions were analyzed using the McPAS-TCR and VDJ databases. TCR clustering was implemented using Grouping of Lymphocyte Interactions by Paratope Hotspots software. Patients with poor prognosis had ascites characterized by an increased ratio of CD8+ T cells to regulatory T cells, which correlated with an increased productive frequency of the top 100 clones and decreased productive entropy. TCRs enriched in patients with an excellent or good prognosis were more likely to recognize cancer antigens and contained more TCR reads predicted to recognize epithelial ovarian cancer antigens. In addition, a TCR motif that binds the TP53 neoantigen was identified, and this motif was enriched in patients with an excellent or good prognosis. Ascitic fluid in high-grade epithelial ovarian cancer patients with an excellent or good prognosis is enriched with TCRs that may recognize ovarian cancer-specific neoantigens, including mutated TP53 and TEAD1. These results suggest that an effective antigen-specific immune response in ascites is vital for a good outcome in high-grade epithelial ovarian cancer.","prettyUrl":"court-2023-po","following":false,"created":"12/15/2022","featured":false,"publishedDate":"12/15/2022","urlOrId":"court-2023-po","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"5c81716a-aab5-496f-9f9c-85e73a82f500","title":"Clonal composition and persistence of antigen-specific circulating T follicular helper cells","investigator":"Sallusto, Federica","investigatorInstitution":"Institute for Research in Biomedicine, Università della Svizzera italiana","publicationName":"European Journal of Immunology","researchArea":"Basic Immunology","prettyUrls":{"273":"hu-2022-eji"},"prettyUrlList":["hu-2022-eji"],"summary":"T follicular helper (TFH) cells are localized in secondary lymphoid organs and play an essential role in promoting B cell responses and antibody affinity maturation in germinal centres (GC). A subset of memory CD4+ T cells expressing the TFH-signature chemokine receptor CXCR5 has been described in human blood as being phenotypically and clonally related to GC TFH. However, the antigen specificity and the relationship of these circulating TFH (cTFH) cells with other circulating memory CD4+ T cells remain poorly defined. In this study, we combined antigenic stimulation and high-throughput T cell receptor (TCR) Vβ sequencing to define the repertoire of human memory cTFH cells specific for different microbial antigens. We found that CXCR5+ cTFH subsets comprised cells that responded to tetanus toxoid (TT), influenza vaccine (Flu) or Candida albicans (C.alb) but endowed with different effector functions compared to cells in CXCR5– non-cTFH subsets. Interestingly, cTFH and non-cTFH cells specific for C.alb or TT had a largely overlapping TCR Vβ repertoire while the repertoire of cTFH and non-cTFH cells specific for Flu was distinct. Furthermore, in-depth immune phenotyping showed that Flu-specific but not C.alb-specific PD-1+ cTFH cells had a “GC TFH-like” phenotype, with overexpression of IL21, CXCL13, BCL6. Longitudinal analysis of multi-year serial blood donations showed that Flu-specific cTFH and non-cTFH cells persisted as phenotypically stable repertoires for years. Collectively, our study contributes to the understanding of the relationship of cTFH with non-cTFH cells and provide insights on the heterogeneous origin and persistence of human cTFH cells specific for different antigens.","prettyUrl":"hu-2022-eji","following":false,"created":"10/19/2022","featured":false,"publishedDate":"12/05/2022","urlOrId":"hu-2022-eji","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"066a7d2f-577d-408f-9e51-5e01589d1dea","title":"T cell repertoire profiling in allografts and native tissues in recipients with COVID-19 after solid organ transplantation: insight into T cell-mediated allograft protection from viral infection","investigator":"Weiner, Joshua","investigatorInstitution":"Columbia University Irving Medical Center","publicationName":"Frontiers in Immunology","researchArea":"Organ transplant","prettyUrls":{"272":"fu-2022-fi"},"prettyUrlList":["fu-2022-fi"],"summary":"The effects of the SARS-CoV-2 virus on the body, and why the effects are more severe in certain patients, remain incompletely understood. One population of special interest is transplant recipients because of their immunosuppressed state. Understanding the pathophysiology of graft dysfunction in transplant patients with the COVID-19 viral syndrome is important for prognosticating the risk to the graft as well as understanding how best to prevent and, if necessary, treat graft injury in these patients. We analyzed multiple types of solid organ transplant recipients (liver, kidney, heart or lung) at our institution who died from SARS-CoV-2 and underwent autopsy (n = 6) or whose grafts were biopsied during active SARS-CoV-2 infection (n = 8). Their serum inflammatory markers were examined together with the histological appearance, viral load, and TCR repertoire of their graft tissue and, for autopsy patients, several native tissues. Histology and clinical lab results revealed a systemic inflammatory pattern that included elevated inflammatory markers and diffuse tissue damage regardless of graft rejection. Virus was detected throughout all tissues, although most abundant in lungs. The TCR repertoire was broadly similar throughout the tissues of each individual, with greater sharing of dominant clones associated with more rapid disease course. There was no difference in viral load or clonal distribution of overall, COVID-associated, or putative SARS-CoV-2-specific TCRs between allograft and native tissue. We further demonstrated that SARS-CoV-2-specific TCR sequences in transplant patients lack a donor HLA-restricted pattern,\nregardless of distribution in allograft or native tissues, suggesting that recognition of viral antigens on infiltrating recipient cells can effectively trigger host T cell anti-viral responses in both the host and graft. Our findings suggest a systemic immune response to the SARS-CoV-2 virus in solid organ transplant patients that is not associated with rejection and consistent with a largely destructive effect of recipient HLA-restricted T cell clones that affects donor and native organs similarly.","prettyUrl":"fu-2022-fi","following":false,"created":"11/18/2022","featured":false,"publishedDate":"12/01/2022","urlOrId":"fu-2022-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"8f249443-8973-4f80-b9fe-5da8fc58796c","title":"Immunopathological signatures in multisystem inflammatory syndrome in children and pediatric COVID-19","investigator":"Notarangelo, Luigi","investigatorInstitution":"Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health","publicationName":"Nature Medicine","researchArea":"Infectious Disease","prettyUrls":{"271":"sacco-2021-misc"},"prettyUrlList":["sacco-2021-misc"],"summary":"Pediatric Coronavirus Disease 2019 (pCOVID-19) is rarely severe; however, a minority of children infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) might develop multisystem inflammatory syndrome in children (MIS-C), with substantial morbidity. In this longitudinal multi-institutional study, we applied multi-omics (analysis of soluble biomarkers, proteomics, single-cell gene expression and immune repertoire analysis) to profile children with COVID-19 (n = 110) and MIS-C (n = 76), along with pediatric healthy controls (pHCs; n = 76). pCOVID-19 was characterized by robust type I interferon (IFN) responses, whereas prominent type II IFN-dependent and NF-κB-dependent signatures, matrisome activation and increased levels of circulating spike protein were detected in MIS-C, with no correlation with SARS-CoV-2 PCR status around the time of admission. Transient expansion of TRBV11-2 T cell clonotypes in MIS-C was associated with signatures of inflammation and T cell activation. The association of MIS-C with the combination of HLA A*02, B*35 and C*04 alleles suggests genetic susceptibility. MIS-C B cells showed higher mutation load than pCOVID-19 and pHC. These results identify distinct immunopathological signatures in pCOVID-19 and MIS-C that might help better define the pathophysiology of these disorders and guide therapy.","prettyUrl":"sacco-2021-misc","following":false,"created":"11/02/2022","featured":false,"publishedDate":"11/29/2022","urlOrId":"sacco-2021-misc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"ecf9a1fc-611d-47dd-a4ba-31cf541444b3","title":"A TLR7-nanoparticle adjuvant promotes broad immune responses against heterologous strains of Influenza and SARS-CoV-2","investigator":"Pulendran, Bali","investigatorInstitution":"Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University","publicationName":"Nature Materials","researchArea":"Vaccine efficacy","prettyUrls":{"270":"yin-2022-nm"},"prettyUrlList":["yin-2022-nm"],"summary":"Effective vaccines against viruses such as Influenza and SARS-CoV-2 must elicit a diverse repertoire of antibodies against multiple variant virus strains. However, antibody responses to current vaccines often lack cross-reactivity due to immunodominance. Here, we describe a polymeric toll-like receptor 7 agonist -nanoparticle adjuvant, TLR7-NP, which enhances lymph node targeting, and leads to persistent activation of immune cells and broad immune responses. When mixed with Alum-adsorbed antigens, this TLR7-NP adjuvant elicited cross-reactive antibodies for both dominant and subdominant epitopes and antigen-specific CD8+ T cell responses in mice. TLR7-NP adjuvanted influenza subunit vaccine successfully protects mice from heterologous viral challenge. TLR7-NP also enhances the antibody response to a SARS-CoV-2 subunit vaccine against multiple variants and mobilizes antiviral responses. Moreover, TLR7-NP augments antigen-specific responses in human tonsil organoids. Overall, we describe a nanoparticle adjuvant to improve the immune response to viral antigens, with promising implications for vaccine development.","prettyUrl":"yin-2022-nm","following":false,"created":"11/07/2022","featured":false,"publishedDate":"11/22/2022","urlOrId":"yin-2022-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"bd1d9a58-8cd6-4ef1-bd67-377d748f3635","title":"Characterization of T Cell Receptor Repertoires in Celiac Disease","investigator":"Bhagat, Govind","investigatorInstitution":"Department of Pathology and Cell Biology, Columbia University","publicationName":"Journal of Clinical Pathology","researchArea":"Gastrointestinal","prettyUrls":{"269":"lee-2022-jcp"},"prettyUrlList":["lee-2022-jcp"],"summary":"Aims: Characterize T-cell receptor gene (TR) repertoires of small intestinal T cells of patients with newly diagnosed (active) celiac disease (ACD), refractory celiac disease type I (RCD I) and CD patients on a gluten-free diet (GFD).\n\nMethods: Next-generation sequencing of complementarity-determining region 3 (CDR3) of rearranged T cell receptor beta (TRB) and gamma (TRG) genes was performed using DNA extracted from intraepithelial cell (IEC) and lamina propria cell (LPC) fractions and a small subset of peripheral blood mononuclear cell (PBMC) samples obtained from CD and non-CD (control) patients. Several parameters were assessed, including relative abundance and enrichment.\n\nResults: TRB and TRG repertoires in CD tissue samples demonstrated lower clonality but higher frequency of rearranged TRs compared to controls. No CD-related differences were detected in the limited number of PBMC samples. Previously published LP gliadin-specific TRB sequences were more frequently detected in LPC samples from CD patients compared to non-CD controls. TRG repertoires of IECs from both ACD and GFD patients demonstrated increased abundance of certain amino acid (AA) motifs compared to controls, encoded by multiple nucleotide variants, including one motif that was enriched in duodenal IECs versus the blood of CD patients.\n\nConclusions: Small intestinal TRB and TRG repertoires of CD patients are more diverse than individuals without CD, likely due to mucosal recruitment and accumulation of T cells because of protracted inflammation. Enrichment of the unique TRG CDR3 AA sequence in the mucosa of CD patients may suggest disease-associated changes in the TCRGD IE lymphocyte (IEL) landscape.","prettyUrl":"lee-2022-jcp","following":false,"created":"09/29/2021","featured":false,"publishedDate":"11/18/2022","urlOrId":"lee-2022-jcp","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"52914af5-01b0-449e-9c6d-a5ee422cb08e","title":"A phase I/Ib trial and analysis of biological correlates of response to neoadjuvant SBRT with single-dose durvalumab in HPV-unrelated locally advanced HNSCC","investigator":"Darragh, Laurel","investigatorInstitution":"Colorado University","publicationName":"Nature Cancer","researchArea":"Cancer Immunotherapy","prettyUrls":{"268":"darragh-2022-nc"},"prettyUrlList":["darragh-2022-nc"],"summary":"Five-year survival for HPV-unrelated head and neck squamous cell carcinomas remains below 50%. We assessed the safety of administering combination hypofractionated stereotactic body radiation therapy (SBRT) with single-dose Durvalumab (anti-PDL-1) neoadjuvantly (n=21) (NCT03635164). The primary endpoint of the study was safety, which was met. Secondary endpoints included radiographic, pathologic, and objective response, locoregional control (LRC), progression-free survival (PFS), and overall survival (OS). Among evaluable patients at an early median follow-up of 16 months (448 days, or 64 weeks), OS was 80.1% with 95% C.I. (62.0%, 100.0%), LRC and PFS were 75.8% with 95% C.I. (57.5%, 99.8%), and major pathological response (MPR) or complete response (CR) was 75% with 95% exact C.I. (51.6%, 100%). Using high-dimensional multi-omics and spatial data as well as biological correlatives, we show that responders had: 1) an increase in effector T cells; 2) a decrease in immunosuppressive cells; and 3) an increase in antigen presentation post-treatment.","prettyUrl":"darragh-2022-nc","following":false,"created":"08/29/2022","featured":false,"publishedDate":"11/02/2022","urlOrId":"darragh-2022-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"6a8d5109-613a-4236-9cd1-764a5b2cf049","title":"TCR repertoire analysis of BCMAxCD3 T cell Engagers (TCE)-treated multiple myeloma","investigator":"Friedrich, Mirco Julian","investigatorInstitution":"Broad Institute of Harvard & MIT","publicationName":"Cancer Cell","researchArea":"Cancer Immunotherapy","prettyUrls":{"267":"friedrich-2022-cc"},"prettyUrlList":["friedrich-2022-cc"],"summary":"Adaptive immunotherapy including bispecific T cell engagers (TCE) has shown promise in the treatment of various cancers, but clinical responses are heterogeneous and often not durable. As response to TCE is suggested to be independent of tumor recognition and T cell state, molecular determinants or mediators of primary and acquired resistance towards TCE remain poorly understood. \n\nHere, we identify conserved behaviors of bone marrow residing CD4+ and CD8+ T cells in multiple myeloma patients undergoing TCE therapy. We show that the bone marrow immune landscape reacts to TCE therapy with cell state-dependent clonal T cell expansion and herewith infer coupling of tumor recognition via MHC-I, T cell exhaustion and clinical response. While we find the abundance of exhausted-like memory CD8+ T cell clones to be associated with clinical response failure, we describe loss of target epitope and MHC class I expression as tumor-intrinsic mechanisms of acquired resistance to TCE.","prettyUrl":"friedrich-2022-cc","following":false,"created":"10/26/2022","featured":false,"publishedDate":"10/28/2022","urlOrId":"friedrich-2022-cc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"439be9ea-89b7-4201-8e66-5e526ba0ad30","title":"The ectonucleotidase CD39 identifies tumor- reactive CD8+ T cells predictive of immune checkpoint blockade efficacy in human lung cancer","investigator":"Chow, Andrew","investigatorInstitution":"Memorial Sloan Kettering Cancer Center","publicationName":"Immunity","researchArea":"Cancer","prettyUrls":{"266":"chow-2022-i"},"prettyUrlList":["chow-2022-i"],"summary":"Improved identification of anti-tumor T cells is needed to advance cancer immunotherapies. CD39 expression is a promising surrogate of tumor-reactive CD8+ T cells. Here, we comprehensively profiled CD39 expression in human lung cancer. CD39 expression enriched for CD8+ T cells with features of exhaustion, tumor reactivity, and clonal expansion. Flow cytometry of 440 lung cancer biospecimens revealed weak association between CD39+ CD8+ T cells and tumoral features, such as programmed death-ligand 1 (PD-L1), tumor mutation burden, and driver mutations. Immune checkpoint blockade (ICB), but not cytotoxic chemotherapy, increased intratumoral CD39+ CD8+ T cells. Higher baseline frequency of CD39+ CD8+ T cells conferred improved clinical outcomes from ICB therapy. Furthermore, a gene signature of CD39+ CD8+ T cells predicted benefit from ICB, but not chemotherapy, in a phase III clinical trial of non-small cell lung cancer. These findings highlight CD39 as a proxy of tumor-reactive CD8+ T cells in human lung cancer.","prettyUrl":"chow-2022-i","following":false,"created":"10/17/2022","featured":false,"publishedDate":"10/28/2022","urlOrId":"chow-2022-i","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"27a5b01f-0e44-4c70-b5fd-730d60ca7b94","title":"Cetuximab responses in HNSCC patients correlate to clonal expansion feature of peripheral and tumor-infiltrating T cells with top T cell receptor (TCR) clonotypes","investigator":"Ferris, Robert","investigatorInstitution":"UPMC Hillman Cancer Center, Department of Otolaryngology, University of Pittsburg","publicationName":"Clinical Cancer Research","researchArea":"Basic Immunology","prettyUrls":{"265":"ge-2022-ccr"},"prettyUrlList":["ge-2022-ccr"],"summary":"Purpose: Cetuximab is a standard-of-care treatment for head and neck squamous cell carcinoma (HNSCC). Well-defined correlative markers of therapeutic responses are still lacking. Characterizing dynamic changes of T cell receptor (TCR) repertoire in peripheral blood and tumor tissue may facilitate developing markers for cetuximab response in HNSCCs.\n\nExperimental Design: We analyzed high-throughput TCR sequencing data generated with ImmunoSEQ platform using peripheral blood and tumor infiltrating lymphocytes (TIL) from HNSCC patients before and after cetuximab treatment (pre-/post-PBMC vs. pre-/post-TIL). Multiple analytic approaches were employed to normalize sequencing data.\n\nResults: Normalized TCR richness was significantly lower in post-TIL than pre-TIL, suggesting that cetuximab reduced TCR diversity and promoted TCR expansion in TIL samples, regardless of response status. The magnitude of clonal expansion (defined as expansion rate) in top-20 TCR clonotypes was significantly higher in responder PBMC with or without normalization, and in responder TIL upon normalization, than non-responder ones. Notably, the expanded top-20 or top-50 TCR clonotypes overlapped between PBMC and TIL samples, which occurred significantly more frequently in responders than non-responders.\n\nConclusions: Cetuximab-treated HNSCC patients harbor dynamic changes of TCR repertoires correlative to therapeutic responses. The expansion rate of top TCR clonotypes in peripheral blood may serve as a minimally invasive, readily accessible, and feasible marker for predicting cetuximab responses in HNSCCs and beyond, and the expansion rate of top TCR clonotypes in TILs and their overlapping probability between PBMC and TIL may serve as additional predictive markers. Our study also highlights the importance of data normalization for TCR repertoire analysis.","prettyUrl":"ge-2022-ccr","following":false,"created":"10/12/2022","featured":false,"publishedDate":"10/26/2022","urlOrId":"ge-2022-ccr","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"58f80131-b5c4-44cf-b07a-ff75d34ee2f2","title":"T cell receptor repertoires associated with control and disease progression following Mycobacterium tuberculosis infection","investigator":"Scriba, Thomas J.","investigatorInstitution":"South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town","publicationName":"Nature Medicine","researchArea":"Infectious Disease","prettyUrls":{"264":"musvosvi-2022-nm"},"prettyUrlList":["musvosvi-2022-nm"],"summary":"Antigen-specific, MHC-restricted ab T cells are necessary for protective immunity against Mycobacterium tuberculosis (M.tb), but the ability to broadly study these responses has been limited. Here we used single cell and bulk TCR sequencing and the GLIPH2 algorithm to analyze millions of M.tb-specific sequences in two longitudinal cohorts, comprising 166 individuals with M.tb infection who either progressed to tuberculosis (N = 48) or controlled infection (N = 118). We found 24 T cell groups with similar TCRb sequences, predicted by GLIPH2 to have common TCR specificities, which associated with control of infection, and others that associated with progression to disease. Using a genome-wide M.tb antigen screen, we identified peptides targeted by T cell similarity groups enriched either in controllers or progressors. We propose that antigens recognized by T cell similarity groups associated with control of infection can be considered high-priority targets for future vaccine development.","prettyUrl":"musvosvi-2022-nm","following":false,"created":"10/17/2022","featured":false,"publishedDate":"10/20/2022","urlOrId":"musvosvi-2022-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"cf68c71a-abd0-452a-b036-7a052a907a5a","title":"Selective retention of virus-specific tissue-resident T cells in healed skin after recovery from herpes zoster","investigator":"Laing, Kerry J","investigatorInstitution":"Department of Medicine, University of Washington","publicationName":"Nature Communications","researchArea":"Infectious Disease","prettyUrls":{"263":"laing-2022-nc"},"prettyUrlList":["laing-2022-nc"],"summary":"Herpes zoster is a localized skin infection caused by reactivation of latent varicella-zoster virus. Tissue-resident T cells likely control skin infections. Zoster provides a unique opportunity to determine if focal reinfection of human skin boosts local or disseminated antigen-specific tissue-resident T cells. Here, we show virus-specific T cells are retained over one year in serial samples of rash site and contralateral unaffected skin of individuals recovered from zoster. Consistent with zoster resolution, viral DNA is largely undetectable on skin from day 90 and virus-specific B and T cells decline in blood. In skin, there is selective infiltration and long-term persistence of varicella-zoster virus-specific T cells in the rash site relative to the contralateral site. The skin T cell infiltrates express the canonical tissue-resident T cell markers CD69 and CD103. These findings show that zoster promotes spatially-restricted long-term retention of antigen-specific tissue-resident T cells in previously infected skin.","prettyUrl":"laing-2022-nc","following":false,"created":"07/21/2022","featured":false,"publishedDate":"09/29/2022","urlOrId":"laing-2022-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"41ccd59e-b478-499e-bb7e-e03cfa88b76f","title":"Clinical relevance of the combined analysis of circulating tumor cells and anti-tumor T-cell immunity in metastatic breast cancer patients","investigator":"Brisotto, Giulia","investigatorInstitution":"Immunopathology and Cancer Biomarkers Units, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO)","publicationName":"Frontiers in Oncology","researchArea":"Cancer","prettyUrls":{"262":"muraro-2022-fo"},"prettyUrlList":["muraro-2022-fo"],"summary":"Background: Metastatic breast cancer (mBC) is a heterogeneous disease with varying responses to treatments and clinical outcomes, still requiring the identification of reliable predictive biomarkers. In this context, liquid biopsy has emerged as a powerful tool to assess in real-time the evolving landscape of cancer, which is both orchestrated by the metastatic process and immune-surveillance mechanisms. Thus, we investigated circulating tumor cells (CTCs) coupled with peripheral T-cell immunity to uncover their potential clinical relevance in mBC.\nMethods: A cohort of 20 mBC patients was evaluated, before and one month after starting therapy, through the following liquid biopsy approaches: CTCs enumerated by a metabolism-based assay, T-cell responses against tumor-associated antigens (TAA) characterized by interferon-γ enzyme-linked immunosorbent spot (ELISpot), and the T-cell receptor (TCR) repertoire investigated by a targeted next-generation sequencing technique. TCR repertoire features were characterized by the Morisita’s overlap and the Productive Simpson Clonality indexes, and the TCR richness. Differences between groups were calculated by Fisher’s, Mann-Whitney or Kruskal-Wallis test, as appropriate. Prognostic data analysis was estimated by Kaplan-Meier method.\nResults: Stratifying patients for their prognostic level of 6 CTCs before therapy, TAA specific T-cell responses were detected only in patients with a low CTC level. By analyzing the TCR repertoire, the highest TCR clonality was observed in the case of CTCs under the cut-off and a positive ELISpot response (p=0.03). Whereas, at follow-up, patients showing a good clinical response coupled with a low number of CTCs were characterized by the most elevated TCR clonality (p<0.05). The detection of CTCs≥6 in at least one time-point was associated with a lower TCR clonality (p=0.02). Intriguingly, by combining overall survival analysis with TCR repertoire, we highlighted a potential prognostic role of the TCR clonality measured at follow-up (p=0.03). \nConclusion: These data, whether validated in a larger cohort of patients, suggest that the combined analysis of CTCs and circulating anti-tumor T-cell immunity could represent a valuable immune-oncological biomarker for the liquid biopsy field. The clinical application of this promising tool could improve the management of mBC patients, especially in the setting of immunotherapy, a rising approach for BC treatment requiring reliable predictive biomarkers.","prettyUrl":"muraro-2022-fo","following":false,"created":"08/04/2022","featured":false,"publishedDate":"09/20/2022","urlOrId":"muraro-2022-fo","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"0e747ae4-093a-4754-bd7b-c0cd31f8b3f7","title":"T Cell Receptor Sequencing Reveals Reduced Clonal Breadth of T Cell Responses against SARS-CoV-2 after Natural Infection and Vaccination in Allogeneic Hematopoietic Stem Cell Transplant Recipients","investigator":"Simonetta, Federico","investigatorInstitution":"Geneva University Hospitals and University of Geneva , Division of Hematology, Department of Oncology","publicationName":"Annals of Oncology","researchArea":"BMT / Stem Cell Transplant","prettyUrls":{"261":"pradier-2022-ao"},"prettyUrlList":["pradier-2022-ao"],"summary":"Allogeneic hematopoietic stem cell transplantation (HSCT) is still associated with significant morbidity and mortality related to infectious complications, with viral infections being the most frequent infectious events. Allogenic HSCT recipients have a higher risk to develop severe forms of COVID-19 and a higher mortality rate after infection with SARS-CoV2 compared to the general population. Prevention by vaccination is critically important in allogeneic HSCT recipients but impaired humoral and cellular responses have been reported. To gain further insights into the immune defects leading to impaired immune responses to SARS-CoV-2 in allogeneic HSCT recipients we performed high-throughput T cell receptor (TCR) repertoire profiling of cells recovered from allogeneic HSCT recipients or healthy controls after SARS-CoV2 natural infection or mRNA-based vaccination. \nSARS-CoV-2- specific T cell clonotypes were detectable in both HC (n=10) and HSCT (n=11) recipients after COVID-19 infection. However, HSCT recipients displayed significantly reduced SARS-CoV-2-specific T cell clonotypes and a less diverse TCR repertoire, as revealed by higher Simpson clonality, compared with HC. Performing the same analysis on samples recovered from allogeneic HSCT recipients (n=11) or from healthy controls (n=10) after vaccination with 3 doses of mRNA-based SARS-CoV-2 vaccines, we observed a significant reduction in S-protein-specific T cell clonotypes in allogeneic HSCT recipient compared to HC. We detected a significant negative correlation between the Simpson clonality and the number of SARS-CoV-2-specific T cell clonotypes following natural infection or vaccination.\nOur results indicate that allogeneic HSCT recipients display a defect in cellular SARS-CoV-2-specific responses after COVID-19 infection and vaccination. Such impairment was associated with increased T-cell clonality after HSCT, pointing to the reduced diversity of the TCR repertoire as a mechanism leading to impaired cellular responses against SARS-CoV-2 in HSCT recipients. SARS-CoV-2 infection represents an unprecedented opportunity to study the immune responses established in allogeneic HSCT recipients against a new pathogen that either the donor or the HSCT recipient have never been exposed to. Our findings provide insights into our understanding of immune-dysfunction after allogeneic HSCT.","prettyUrl":"pradier-2022-ao","following":false,"created":"08/30/2022","featured":false,"publishedDate":"09/16/2022","urlOrId":"pradier-2022-ao","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a0151bae-347f-4fa6-9e8d-252733f8b6cb","title":"Evolution and modulation of antigen-specific T cell responses in melanoma patients","investigator":"Huuhtanen, Jani","investigatorInstitution":"Hematology Research Unit Helsinki, University of Helsinki","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"260":"huuhtanen-2022-nc"},"prettyUrlList":["huuhtanen-2022-nc"],"summary":"Analyzing antigen-specific T cell responses at scale has been challenging. Here, we analyze three types of T cell receptor (TCR) repertoire data (antigen-specific TCRs, TCR-repertoire, and single-cell RNA+TCRαβ-sequencing data) from 515 patients with primary or metastatic melanoma and compare it to 783 healthy controls. Although melanoma-associated antigen (MAA) -specific TCRs are restricted to individuals, they share sequence similarities that allow us to build classifiers for predicting anti-MAA T cells. The frequency of anti-MAA T cells distinguishes melanoma patients from healthy and predicts metastatic recurrence from primary melanoma. Anti-MAA T cells have stem-like properties and frequent interactions with regulatory T cells and tumor cells via Galectin9-TIM3 and PVR-TIGIT -axes, respectively. In the responding patients, the number of expanded anti-MAA clones are higher after the anti-PD1(+anti-CTLA4) therapy and the exhaustion phenotype is rescued. Our systems immunology approach paves the way for understanding antigen-specific responses in human disorders.","prettyUrl":"huuhtanen-2022-nc","following":false,"created":"08/23/2022","featured":false,"publishedDate":"09/12/2022","urlOrId":"huuhtanen-2022-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"f29f5aa5-f20a-404f-901d-bf29d8ab1bf1","title":"Siglec-7 represents a glyco-immune checkpoint for non-exhausted effector memory CD8+ T cells with high functional and metabolic capacities","investigator":"von Guten, Stephan","investigatorInstitution":"Institute of Pharmacology, University of Bern","publicationName":"Frontiers in Immunology","researchArea":"Basic Immunology","prettyUrls":{"259":"haas-2022-fi"},"prettyUrlList":["haas-2022-fi"],"summary":"While inhibitory Siglec receptors are known to regulate myeloid cells, less is known about their expression and function in lymphocytes subsets. Here we identified Siglec-7 as a lycol-immune checkpoint expressed on non-exhausted effector memory CD8+ T cells that exhibit high functional and metabolic capacities. Seahorse analysis revealed higher basal respiration and glycolysis levels of Siglec-7+ CD8+ T cells in steady state, and particularly upon activation. Siglec-7 polarization into the T cell immune synapse was dependent on sialoglycan interactions in trans and prevented actin polarization and effective T cell responses. Siglec-7 ligands were found to be expressed on both leukemic stem cells and acute myeloid leukemia (AML) cells suggesting the occurrence of lycol-immune checkpoints for Siglec-7+ CD8+ T cells, which were found in patients’ peripheral blood and bone marrow. Our findings project Siglec-7 as a lycol-immune checkpoint and therapeutic target for T cell-driven disorders and cancer.","prettyUrl":"haas-2022-fi","following":false,"created":"08/19/2022","featured":false,"publishedDate":"09/06/2022","urlOrId":"haas-2022-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"4971360b-869c-45e5-9364-1f6cecce1355","title":"Anti-spike T-cell and Antibody Responses to SARS-CoV-2 mRNA Vaccines in Patients with Hematologic Malignancies","investigator":"Greenberger, Lee M","investigatorInstitution":"The Leukemia & Lymphoma Society","publicationName":"Blood Cancer Discovery","researchArea":"Infectious Disease","prettyUrls":{"258":"greenberger-2022-bcd"},"prettyUrlList":["greenberger-2022-bcd"],"summary":"The anti-spike T-cell and antibody responses to SARS-CoV-2 mRNA vaccines in patients with B-cell malignancies were examined in a real-world setting. A next-generation sequencing (NGS)-based molecular assay was used to assess SARS-CoV-2-specific T-cell responses. After the second dose, 58% (166/284) of seropositive and 45% (99/221) of seronegative patients display anti-spike T cells. The percentage of patients who displayed T-cell response was higher among patients receiving mRNA-1273 vaccines compared with those receiving BNT162b2 vaccines. After the third vaccination, 40% (137/342) of patients seroconverted, although only 22% displayed sufficient antibody levels associated with the production of neutralizing antibodies. 97% (717/738) of patients who were seropositive before the third dose had markedly elevated anti-spike antibody levels. Anti-spike antibody levels, but not T-cell responses, were depressed by B cell-directed therapies. Vaccinated patients with B-cell malignancies with a poor response to SARS-CoV-2 vaccines may remain vulnerable to COVID-19 infections.","prettyUrl":"greenberger-2022-bcd","following":false,"created":"08/23/2022","featured":false,"publishedDate":"09/02/2022","urlOrId":"greenberger-2022-bcd","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b1a51655-6b1a-4eb2-a349-2aeba44863b0","title":"SARS-CoV-2-Specific T Cell Responses in Patients with Multisystem Inflammatory Syndrome in Children","investigator":"Henderson, Lauren A","investigatorInstitution":"Division of Immunology, Boston Children’s Hospital, Harvard Medical School","publicationName":"Clinical Immunology","researchArea":"Infectious Disease","prettyUrls":{"257":"lam-2022-ci"},"prettyUrlList":["lam-2022-ci"],"summary":"Multisystem inflammatory syndrome in children (MIS-C) is a severe complication of SARS-CoV-2 infections that occurs in the pediatric population. We sought to characterize T cell responses in MIS-C compared to COVID-19 and pediatric hyperinflammatory syndromes. MIS-C was distinct from COVID-19 and hyperinflammatory syndromes due to an expansion of T cells expressing TRBV11-2 that was not associated with HLA genotype. Children diagnosed with MIS-C, but who were negative for SARS-CoV-2 by PCR and serology, did not display Vβ skewing. There was no difference in the proportion of T cells that became activated after stimulation with SARS-CoV-2 peptides in children with MIS-C compared to convalescent COVID-19. The frequency of SARS-CoV-2-specific TCRs and the antigens recognized by these TCRs were comparable in MIS-C and COVID-19. Expansion of Vβ11-2+ T cells was a specific biomarker of MIS-C patients with laboratory confirmed SARS-CoV-2 infections. Children with MIS-C had robust antigen-specific T cell responses to SARS-CoV-2.","prettyUrl":"lam-2022-ci","following":false,"created":"08/30/2022","featured":false,"publishedDate":"08/30/2022","urlOrId":"lam-2022-ci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"804308d7-33d4-4017-a125-8d77143998d9","title":"Autoreactive napsin A-specific T cells are enriched in lung tumors and inflammatory lung lesions during immune checkpoint blockade","investigator":"Flatz, Lukas","investigatorInstitution":"Department of Dermatology, University Hospital Tübingen","publicationName":"Science Immunology","researchArea":"Cancer Immunotherapy","prettyUrls":{"256":"berner-2022-si"},"prettyUrlList":["berner-2022-si"],"summary":"Cancer treatment with immune checkpoint blockade (ICB) often induces immune-related adverse events (irAEs). We hypothesized that proteins coexpressed in tumors and normal cells could be antigenic targets in irAEs and herein described DITAS (discovery of tumor-associated self-antigens) for their identification. DITAS computed transcriptional similarity between lung tumors and healthy lung tissue based on single-sample gene set enrichment analysis. This identified 10 lung tissue-specific genes highly expressed in the lung tumors. Computational analysis was combined with functional T cell assays and single-cell RNA sequencing of the antigen-specific T cells to validate the lung tumor self-antigens. In patients with non-small cell lung cancer (NSCLC) treated with ICB, napsin A was a self-antigen that elicited strong CD8+ T cell responses, with ICB responders harboring higher frequencies of these CD8+ T cells compared with nonresponders. Human leukocyte antigen (HLA) class I ligands derived from napsin A were present in human lung tumors and in nontumor lung tissues, and napsin A tetramers confirmed the presence of napsin A-specific CD8+ T cells in blood and tumors of patients with NSCLC. Napsin A-specific T cell clonotypes were enriched in lung tumors and ICB-induced inflammatory lung lesions and could kill immortalized HLA-matched NSCLC cells ex vivo. Single-cell RNA sequencing revealed that these T cell clonotypes expressed proinflammatory cytokines and cytotoxic markers. Thus, DITAS successfully identified self-antigens, including napsin A, that likely mediate effective antitumor T cell responses in NSCLC and may simultaneously underpin lung irAEs.","prettyUrl":"berner-2022-si","following":false,"created":"08/08/2022","featured":false,"publishedDate":"08/11/2022","urlOrId":"berner-2022-si","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"5d8d03a1-b6e5-44d6-b939-81e1bf36e29c","title":"Temporal development of T cell receptor repertoires during childhood in health and disease","investigator":"MIchels, Aaron W.","investigatorInstitution":"Barbara Davis Center for Diabetes, University of Colorado School of Medicine","publicationName":"JCI Insight","researchArea":"Basic Immunology","prettyUrls":{"255":"mitchell-2022-jcii"},"prettyUrlList":["mitchell-2022-jcii"],"summary":"T cell receptor (TCR) sequences are exceptionally diverse and can now be comprehensively measured with next generation sequencing technologies. However, a thorough investigation of longitudinal TCR repertoires throughout childhood in health and during development of a common childhood disease, type 1 diabetes (T1D), has not been undertaken. Here, we deep-sequenced the TCR beta chain (TCRβ) repertoires from longitudinal peripheral blood DNA samples at four time points beginning early in life (median age of 1.4 years) from children that progressed to T1D (n=29) and age/sex-matched islet autoantibody negative controls (n=25). From 53 million TCRβ sequences, we show that the repertoire is extraordinarily diverse early in life and narrows with age independent of disease. We demonstrate the ability to identify specific TCR sequences, including those known to recognize influenza A and separately those specific for insulin and its precursor, preproinsulin. Insulin-reactive TCRβ sequences are more common and frequent in number as the disease progressed in those who developed T1D compared to genetically at-risk non-diabetic children, which was not the case for influenza-reactive sequences. As an independent validation, we sequenced and analyzed TCRβ repertoires from a cohort of new-onset T1D patients (n=143), identifying the same preproinsulin-reactive TCRs. These results demonstrate an enrichment of preproinsulin-reactive TCR sequences during the progression to T1D, highlighting the importance of using disease-relevant TCR sequences as powerful biomarkers in autoimmune disorders.","prettyUrl":"mitchell-2022-jcii","following":false,"created":"06/15/2022","featured":false,"publishedDate":"07/14/2022","urlOrId":"mitchell-2022-jcii","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"ab25f2b3-f385-4668-9e6c-6369f12c907e","title":"Single-cell profiling of the leukemic and non-leukemic immune cell compartments in CD8+ T-cell Large Granular Lymphocytic Leukemia","investigator":"Huuhtanen, Jani","investigatorInstitution":"Hematology Research Unit Helsinki, University of Helsinki","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"254":"huuhtanen-2021-nc"},"prettyUrlList":["huuhtanen-2021-nc"],"summary":"T-cell large granular lymphocytic leukemia (T-LGLL) is a rare lymphoproliferative disorder of mature, clonally expanded T-cells, where somatic-activating STAT3 mutations are common. Although T-LGLL has been described as a chronic T-cell response to an antigen, the role of the non-leukemic immune system in this response is largely uncharacterized. By utilizing single-cell RNA and T-cell receptor profiling (scRNA+TCRαβ-seq), we show that irrespective of STAT3 mutation status, T-LGLL clonotypes are more cytotoxic and exhausted than healthy reactive clonotypes. In addition, T-LGLL clonotypes show more active cell communication than reactive clones with non-leukemic immune cells via costimulatory cell–cell interactions, monocyte-secreted proinflammatory cytokines, and T-LGLL-clone-secreted IFNγ. Besides the leukemic repertoire, the non-leukemic T-cell repertoire in T-LGLL is also more mature, cytotoxic, and clonally restricted than in other cancers and autoimmune disorders. Finally, 67% of the leukemic T-LGLL clonotypes share T-cell receptor similarities with their non-leukemic repertoire, linking the leukemic and non-leukemic repertoires together via possible common target antigens. Our results provide a rationale to prioritize therapies that target the entire immune repertoire and not only the T-LGLL clonotype.","prettyUrl":"huuhtanen-2021-nc","following":false,"created":"10/08/2021","featured":false,"publishedDate":"05/23/2022","urlOrId":"huuhtanen-2021-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"194fcf49-c500-41ea-addb-0207581af787","title":"Systemic Inflammation and Tumour-Infiltrating T-Cell Receptor Repertoire Diversity Are Predictive of Clinical Outcome in High-Grade B-Cell Lymphoma with MYC and BCL2 and/or BCL6 Rearrangements","investigator":"Gebauer, Niklas","investigatorInstitution":"Department of Haematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck","publicationName":"Cancers","researchArea":"Cancer","prettyUrls":{"253":"olschewski-2021-c"},"prettyUrlList":["olschewski-2021-c"],"summary":"High-grade B-cell lymphoma, with MYC and BCL2 and/or BCL6 rearrangements (double/triple-hit high grade B-cell lymphoma, HGBL-DH/TH) constitutes a provisional entity among B-cell malignancies with an aggressive behavior and dire prognosis. While evidence for the essential prognostic role of the composition of the tumor-microenvironment (TME) in hematologic malignancies is growing, its prognostic impact in HGBL-DH/TH remains unknown. In this study, we outline the adaptive immune response in a cohort of 47 HGBL-DH/TH and 27 triple-negative diffuse large B-cell lymphoma (tnDLBCL) patients in a large-scale, next-generation sequencing (NGS) investigation of the T-cell receptor (TCR) β-chain repertoire and supplement our findings with data on the Glasgow-Prognostic Score (GPS) at diagnosis, as a score-derived measure of systemic inflammation. We supplement these studies with an immunophenotypic investigation of the TME. Our findings demonstrate that the clonal architecture of the TCR repertoire of HGBL-DH/TH differs significantly from tnDLBCL. Moreover, several entity-exclusive clonotypes, suggestive of tumor-neoantigen selection are identified. Additionally, both productive clonality and percentage of maximum frequency clone as measures of TCR repertoire diversity and tumor-directed activity of the adaptive immune system had significant impact on overall survival (OS; productive clonality: p = 0.0273; HR: 2.839; CI: 1.124-7.169; maximum productive frequency: p = 0.0307; HR: 2.167; CI: 1.074-4.370) but not PFS (productive clonality: p = 0.4459; maximum productive frequency: p = 0.5567) in HGBL-DH/TH patients, while GPS was a significant predictor of both OS and PFS (OS: p < 0.0001; PFS: p = 0.0002). Subsequent multivariate analysis revealed GPS and the revised international prognostic index (R-IPI) to be the only prognosticators holding significant impact for OS (GPS: p = 0.038; R-IPI: p = 0.006) and PFS (GPS: p = 0.029; R-IPI: p = 0.006) in HGBL-DH/TH. Through the identification of expanded, recurrent and entity-exclusive TCR-clonotypes we provide indications for a distinct subset of tumor-neoantigenic elements exclusively shared among HGBL-DH/TH. Further, we demonstrate an adverse prognostic role for both systemic inflammation and uniform adaptive immune response.","prettyUrl":"olschewski-2021-c","following":false,"created":"05/20/2022","featured":false,"publishedDate":"05/20/2022","urlOrId":"olschewski-2021-c","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"4ae9ad1c-de36-4acf-80ce-9e799d79abe0","title":"SARS-CoV-2 vaccination diversifies the CD4+ T cell repertoire in patients with prior SARS-CoV-2 infection","investigator":"Smith, Kellie N.","investigatorInstitution":"Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University","publicationName":"EBioMedicine","researchArea":"Vaccine efficacy","prettyUrls":{"252":"dykema-2022-ebiomed"},"prettyUrlList":["dykema-2022-ebiomed"],"summary":"COVID-19 mRNA vaccines elicit strong T and B cell responses to the SARS-CoV-2 spike glycoprotein in both SARS-CoV-2 naïve and experienced patients. However, it is unknown whether the post-vaccine CD4+ T cell boost, critical for virus-specific immune memory, seen in patients with a history of COVID-19 is due to restimulation of T cell clonotypes that were first activated during SARS-CoV-2 primary infection or is the result of new clones that are activated by the vaccine. We identified both new and preexisting T cell receptor clonotypes and found that vaccination significantly broadens the SARS-CoV-2-reactive CD4+ T cell repertoire in patients with a history of COVID-19. Additionally, we demonstrated that the vaccine preferentially induces T cells that only recognize SARS-CoV-2 antigens, rather than the less-avid SARS-CoV-2/common cold coronavirus (CCC) cross-reactive responses. These data demonstrate that SARS-CoV-2 vaccination induces a distinct antigen-specific repertoire relative to natural infection and increases the breadth of the SARS-CoV-2-reactive T cell response.","prettyUrl":"dykema-2022-ebiomed","following":false,"created":"08/17/2021","featured":false,"publishedDate":"05/20/2022","urlOrId":"dykema-2022-ebiomed","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"9901707c-4881-4543-b0da-6167ed370e6b","title":"Longitudinal analysis of T-cell receptor repertoires reveals shared patterns of antigen-specific response to SARS-CoV-2 infection","investigator":"Tonon, Giovanni","investigatorInstitution":"IRCCS Ospedale San Raffaele","publicationName":"Journal of Clinical Investigation","researchArea":"Infectious Disease","prettyUrls":{"251":"gittelman-2022-jci"},"prettyUrlList":["gittelman-2022-jci"],"summary":"T cells play a prominent role in orchestrating the immune response to viral diseases, but their role in the clinical presentation and subsequent immunity to SARS-CoV-2 infection remains poorly understood. As part of a population-based survey of the municipality of Vo’, Italy conducted after the initial SARS-CoV-2 outbreak, we sampled the T-cell receptor (TCR) repertoires of the population 2 months after the initial PCR survey and followed up positive cases 9 and 15 months later. At two months, we found that 97.0% (98/101) of cases had elevated levels of TCRs associated with SARS-CoV-2. T-cell frequency (depth) was increased in individuals with more severe disease. Both depth and diversity (breadth) of the TCR repertoire were positively associated with neutralizing antibody titers, driven mostly by CD4+ T cells directed against spike protein. At the later time points, detection of these TCRs remained high, with 90.7% (78/96) and 86.2% (25/29) of individuals having detectable signal at 9 and 15 months, respectively. Forty-three individuals were vaccinated by month 15 and showed a significant increase in TCRs directed against spike protein. Taken together, these results demonstrate the central role of T cells in mounting an immune defense against SARS-CoV-2 that persists out to 15 months.","prettyUrl":"gittelman-2022-jci","following":false,"created":"05/05/2022","featured":false,"publishedDate":"05/16/2022","urlOrId":"gittelman-2022-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1f83aaee-5801-4264-bbae-45d3b56e4904","title":"Microenvironmental correlates of immune checkpoint inhibitor response in human melanoma brain metastases revealed by T cell receptor and single-cell RNA sequencing","investigator":"Brastianos, Priscilla","investigatorInstitution":"Massachusetts General Hospital","publicationName":"Cancer Immunology Research","researchArea":"Cancer Immunotherapy","prettyUrls":{"250":"alvarez-breckenridge-2022-cir"},"prettyUrlList":["alvarez-breckenridge-2022-cir"],"summary":"Melanoma-derived brain metastases (MBM) represent an unmet clinical need due to central nervous system (CNS) progression as a frequent, end-stage site of disease. Immune checkpoint inhibition (ICI) represents a clinical opportunity against MBM; however, the MBM tumor microenvironment (TME) has not been fully elucidated in the context of ICI. To dissect unique MBM-TME elements and correlates of MBM-ICI response, we collected 27 fresh MBM and performed single cell RNA sequencing of the MBM-TME and T cell receptor clonotyping on T cells from MBM and matched blood and extracranial lesions. We observed myeloid phenotypic heterogeneity, most notably multiple distinct neutrophil states including an IL-8 expressing population that correlated with malignant cell epithelial-to-mesenchymal transition. Additionally, we observe significant relationships between intracranial T cell phenotypes and the distribution of T cell clonotypes intracranially and peripherally. We found that the phenotype, clonotype, and overall number of MBM-infiltrating T cells were associated with response to ICI, suggesting that ICI-responsive MBMs interact with peripheral blood in a manner similar to extracranial lesions. These data demonstrate unique features of the MBM-TME, which may represent potential targets to improve clinical outcomes for patients with MBM.","prettyUrl":"alvarez-breckenridge-2022-cir","following":false,"created":"03/19/2022","featured":false,"publishedDate":"05/04/2022","urlOrId":"alvarez-breckenridge-2022-cir","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"9b5be686-453e-4dc1-af85-517d4a5085af","title":"T-cell receptor sequencing identifies prior SARS-CoV-2 infection and correlates with neutralizing antibodies and disease severity","investigator":"Koelle, David M","investigatorInstitution":"University of Washington","publicationName":"JCI insight","researchArea":"Infectious Disease","prettyUrls":{"249":"elyanow-2022-jci"},"prettyUrlList":["elyanow-2022-jci"],"summary":"BACKGROUND. Measuring the immune response to SARS-CoV-2 enables assessment of past infection and protective immunity. SARS-CoV-2 infection induces humoral and T-cell responses, but these responses vary with disease severity and individual characteristics.\n \nMETHODS. A T-cell receptor (TCR) immunosequencing assay was conducted using small-volume blood samples from 302 individuals recovered from COVID-19. Correlations between the magnitude of the T-cell response and neutralizing antibody (nAb) titers or indicators of disease severity were evaluated. Sensitivity of T-cell testing was assessed and compared to serologic testing.\n\nRESULTS. SARS-CoV-2–specific T-cell responses were significantly correlated with nAb titers and clinical indicators of disease severity, including hospitalization, fever, and difficulty breathing. Despite modest declines in depth and breadth of T-cell responses during convalescence, high sensitivity was observed until at least 6 months after infection, with overall sensitivity ~5% greater than serology tests for identifying prior SARS-CoV-2 infection. Improved performance of T-cell testing was most apparent in recovered, non-hospitalized individuals sampled >150 days after initial illness, suggesting greater sensitivity than serology at later timepoints and in individuals with less severe disease. T-cell testing identified SARS-CoV-2 infection in 68% (55/81) of samples with undetectable nAb titers (<1:40) and in 37% (13/35) of samples negative by 3 antibody assays. \n\nCONCLUSION. These results support TCR-based testing as a scalable, reliable measure of past SARS-CoV-2 infection with clinical value beyond serology.","prettyUrl":"elyanow-2022-jci","following":false,"created":"03/31/2022","featured":false,"publishedDate":"04/20/2022","urlOrId":"elyanow-2022-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c06842c7-aab0-4cf7-a298-44745cbea263","title":"PD-1 and ICOS co-expression identifies tumor-reactive CD4 T cells in human solid tumors","investigator":"Duhen, Thomas","investigatorInstitution":"Earle A. Chiles Research Institute, Providence Cancer Institute","publicationName":"Journal of Clinical Investigation","researchArea":"Cancer Immunotherapy","prettyUrls":{"248":"duhen-2022-jci"},"prettyUrlList":["duhen-2022-jci"],"summary":"CD4 T helper (Th) cells play a key role in orchestrating immune responses, but the identity of the CD4 Th cells involved in the anti-tumor immune response remains to be defined. We analyzed the immune cell infiltrates of head and neck squamous cell carcinoma and colorectal cancers and identified a subset of CD4 Th cells distinct from FOXP3+ regulatory T cells that co-expressed PD-1 and ICOS. These tumor-infiltrating CD4 Th cells (CD4 Th TIL) had a tissue-resident memory phenotype, were present in MHC class II-rich areas and proliferated in the tumor suggesting local antigen recognition. The T-cell receptor repertoire of the PD-1+ICOS+ CD4 Th TIL was oligoclonal, with T-cell clones expanded in the tumor, but present at low frequencies in the periphery. Finally, these PD-1+ICOS+ CD4 Th TIL were shown to recognize both tumor-associated antigens and tumor-specific neoantigens. Our findings provide an approach for isolating tumor-reactive CD4 Th TIL directly ex vivo that will help define their role in the anti-tumor immune response and potentially improve future adoptive T-cell therapy approaches.","prettyUrl":"duhen-2022-jci","following":false,"created":"04/18/2022","featured":false,"publishedDate":"04/19/2022","urlOrId":"duhen-2022-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"6c8696a8-e811-4029-a840-88ca2362a974","title":"Neoantigen specific CD4+ T cells in human melanoma have diverse differentiation states and correlate with CD8+ T cell, macrophage, and B cell function","investigator":"Veatch, Joshua","investigatorInstitution":"Fred Hutchinson Cancer Research Center","publicationName":"Cancer Cell","researchArea":"Cancer","prettyUrls":{"247":"veatch-2022-cc"},"prettyUrlList":["veatch-2022-cc"],"summary":"CD4+ T cells that recognize tumor antigens are required for immune checkpoint inhibitor efficacy in murine models, but their contributions in human cancer are unclear. We used single-cell RNA sequencing and T cell receptor sequences to identify signatures and functional correlates of tumor-specific CD4+ T cells infiltrating human melanoma. Conventional CD4+ T cells that recognize tumor neoantigens express CXCL13 and are subdivided into clusters expressing memory and T follicular helper markers, and those expressing cytolytic markers, inhibitory receptors, and IFN-γ. The frequency of CXCL13+ CD4+ T cells in the tumor correlated with the transcriptional states of CD8+ T cells and macrophages, maturation of B cells, and patient survival. Similar correlations were observed in a breast cancer cohort. These results identify phenotypes and functional correlates of tumor-specific CD4+ T cells in melanoma and suggest the possibility of using such cells to modify the tumor microenvironment.","prettyUrl":"veatch-2022-cc","following":false,"created":"03/03/2022","featured":false,"publishedDate":"04/18/2022","urlOrId":"veatch-2022-cc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"3e202bcc-f154-4d79-9f7b-f0524fbc1296","title":"Combinatorial analysis reveals highly coordinated early-stage immune reactions that predict later antiviral immunity in mild COVID-19 patients","investigator":"Ollert, Marcus","investigatorInstitution":"Department of Infection and Immunity, Luxembourg Institute of Health (LIH)","publicationName":"Cell Reports Medicine","researchArea":"Infectious Disease","prettyUrls":{"246":"capelle-2022-crm"},"prettyUrlList":["capelle-2022-crm"],"summary":"While immunopathology has been widely studied in patients with severe COVID-19, immune responses in non-hospitalized patients have remained largely elusive. We systematically analyze 484 peripheral cellular or soluble immune features in a longitudinal cohort of 63 mild and 15 hospitalized patients versus 14 asymptomatic and 26 household controls. We observe a transient increase of IP10/CXCL10 and interferon-β levels, coordinated responses of dominant SARS-CoV-2-specific CD4 and fewer CD8 T cells, and various antigen-presenting and antibody-secreting cells in mild patients within 3 days of PCR diagnosis. The frequency of key innate immune cells and their functional marker expression are impaired in hospitalized patients at day 1 of inclusion. T cell and dendritic cell responses at day 1 are highly predictive for SARS-CoV-2-specific antibody responses after 3 weeks in mild but not hospitalized patients. Our systematic analysis reveals a combinatorial picture and trajectory of various arms of the highly coordinated early-stage immune responses in mild COVID-19 patients.","prettyUrl":"capelle-2022-crm","following":false,"created":"03/15/2022","featured":false,"publishedDate":"04/01/2022","urlOrId":"capelle-2022-crm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"284b2fdc-fc90-4c4e-9807-c248629990eb","title":"Durable expansion of TCR-δ meta-clonotypes after BCG revaccination in humans","investigator":"Seshadri, Chetan","investigatorInstitution":"Department of Medicine, University of Washington","publicationName":"Frontiers in Immunology","researchArea":"Response to Therapeutic Agent","prettyUrls":{"245":"james-2022-fi"},"prettyUrlList":["james-2022-fi"],"summary":"Mycobacterium bovis bacille Calmette-Guérin (BCG) has been used for 100 years and prevents disseminated tuberculosis and death in young children. However, it shows only partial efficacy against pulmonary tuberculosis (TB) in adults, so new vaccines are urgently needed. The protective efficacy of BCG depends on T cells, which are typically activated by pathogen-derived protein antigens that bind to highly polymorphic major histocompatibility complex (MHC) molecules. Some T cells recognize non-protein antigens via antigen presenting systems that are independent of genetic background, leading to their designation as donor-unrestricted T (DURT) cells. Whether live whole cell vaccines, like BCG, can induce durable expansions of DURT cells in humans is not known. We used combinatorial tetramer staining, multi-parameter flow cytometry, and immunosequencing to comprehensively characterize the effect of BCG on activation and expansion of DURT cell subsets. We examined peripheral blood mononuclear cells (PBMC) derived from a Phase I study of South African adults in which samples were archived at baseline, 3 weeks, and 52 weeks post-BCG revaccination. We did not observe a change in the frequency of total mucosal-associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells, germline encoded mycolyl-reactive (GEM) T cells, or T cells at 52 weeks post-BCG. However, immunosequencing revealed a set of TCR- clonotypes that were expanded at 52 weeks post-BCG revaccination. These expanded clones expressed the V2 gene segment and could be further defined on the basis of biochemical similarity into several ‘meta-clonotypes’ that likely recognize similar epitopes. Our data reveal that BCG vaccination leads to durable expansion of DURT cell clonotypes despite a limited effect on total circulating frequencies in the blood and have implications for defining the immunogenicity of candidate whole cell TB vaccines.","prettyUrl":"james-2022-fi","following":false,"created":"03/10/2022","featured":false,"publishedDate":"03/23/2022","urlOrId":"james-2022-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"aaac6745-68ef-437d-bca1-8ccd57d4ec87","title":"Serial analysis of the T-cell repertoire in people living with HIV+ defines the limited impact of long-term anti-retroviral therapy","investigator":"Towlerton, Andrea MH","investigatorInstitution":"Clinical Research Division, Fred Hutchinson Cancer Research Center","publicationName":"Frontiers in Immunology","researchArea":"HIV","prettyUrls":{"244":"towlerton-2022-hiv"},"prettyUrlList":["towlerton-2022-hiv"],"summary":"Long-term antiretroviral therapy (ART) in people living with HIV (PLHIV) is associated with sustained increases in CD4+ T-cell count, but its effect on the peripheral blood T-cell repertoire has not been comprehensively evaluated. In this study, we performed serial profiling of the composition and diversity of the T-cell receptor β-chain (TRB) repertoire in 30 adults with HIV infection before and after the initiation of ART to define its long-term impact on the TRB repertoire. Serially acquired blood samples from 30 adults with HIV infection collected over a mean of 6 years (range, 1-12) years, with 1-4 samples collected before and 2-8 samples collected after the initiation of ART, were available for analysis. TRB repertoires were characterized via high-throughput sequencing of the TRB variable region performed on genomic DNA extracted from unsorted peripheral blood mononuclear cells. Additional laboratory and clinical metadata including serial measurements of HIV viral load and CD4 + T-cell count were available for all individuals in the cohort. A previously published control group of 189 TRB repertoires from peripheral blood samples of adult bone marrow transplant donors was evaluated for comparison. ART initiation in PLHIV was associated with a sustained reduction in viral load and a significant increase in TRB repertoire diversity. However, repertoire diversity in PLHIV remained significantly lower than in the control group even after long-term ART. The composition of TRB repertoires of PLHIV after ART also remained perturbed compared to the control cohort, as evidenced by large persistent private clonal expansions, reduced efficiency in the generation of TRB CDR3 amino acid sequences, and a narrower range of CDR3 lengths. Network analysis revealed an antigen-experienced structure in the TRB repertoire of PLHIV both before and after ART initiation that was quite distinct from the structure of control repertoires, with a slight shift toward a more naïve structure observed after ART initiation. Though we observe significant improvement in TRB repertoire diversity with durable viral suppression in PLHIV on long-term ART, the composition and structure of these repertoires remain significantly perturbed compared to the control cohort of adult bone marrow transplant donors.","prettyUrl":"towlerton-2022-hiv","following":false,"created":"02/18/2022","featured":false,"publishedDate":"03/03/2022","urlOrId":"towlerton-2022-hiv","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d1558b60-30a4-4f50-bbea-cc3a63805d42","title":"Identification of novel STAT5B mutations and characterization of TCRβ signatures in CD4+ T-cell large granular lymphocyte leukemia","investigator":"Bhattacharya, Dipabarna","investigatorInstitution":"Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center","publicationName":"Blood Cancer Journal","researchArea":"Cancer","prettyUrls":{"243":"bhattacharya-2022-bcj"},"prettyUrlList":["bhattacharya-2022-bcj"],"summary":"CD4+ T-cell large granular lymphocyte leukemia (T-LGLL) is a rare subtype of T-LGLL with unknown etiology. In this study, we molecularly characterized a cohort of patients (n = 35) by studying their T-cell receptor (TCR) repertoire and the presence of somatic STAT5B mutations. In addition to the previously described gain-of-function mutations (N642H, Y665F, Q706L, S715F), we discovered six novel STAT5B mutations (Q220H, E433K, T628S, P658R, P702A, and V712E). Multiple STAT5B mutations were present in 22% (5/23) of STAT5B mutated CD4+ T-LGLL cases, either coexisting in one clone or in distinct clones. Patients with STAT5B mutations had increased lymphocyte and LGL counts when compared to STAT5B wild-type patients. TCRβ sequencing showed that, in addition to large LGL expansions, non-leukemic T cell repertoires were more clonal in CD4+ T-LGLL compared to healthy. Interestingly, 25% (15/59) of CD4+ T-LGLL clonotypes were found, albeit in much lower frequencies, in the non-leukemic CD4+ T cell repertoires of the CD4+ T-LGLL patients. Additionally, we further confirmed the previously reported clonal dominance of TRBV6-expressing clones in CD4+ T-LGLL. In conclusion, CD4+ T-LGLL patients have a typical TCR and mutation profile suggestive of aberrant antigen response underlying the disease.","prettyUrl":"bhattacharya-2022-bcj","following":false,"created":"01/12/2022","featured":false,"publishedDate":"02/24/2022","urlOrId":"bhattacharya-2022-bcj","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"880b7ad2-1587-492d-9444-d4366ec71d31","title":"ATR-mediated CD47 and PD-L1 upregulation restricts radiotherapy-induced immune priming and abscopal responses in colorectal cancer","investigator":"Hsieh, Rodney Cheng-En","investigatorInstitution":"Department of Immunology, The University of Texas MD Anderson Cancer Center","publicationName":"Science Immunology","researchArea":"Cancer","prettyUrls":{"242":"hsieh-2022-si"},"prettyUrlList":["hsieh-2022-si"],"summary":"Radiotherapy (RT) of colorectal cancer (CRC) can prime adaptive immunity against tumor-associated antigen (TAA)-expressing CRC cells systemically. However, abscopal tumor remissions are extremely rare, and the postirradiation immune escape mechanisms in CRC remain elusive. Here, we found that irradiated CRC cells used ATR-mediated DNA repair signaling pathway to up-regulate both CD47 and PD-L1, which through engagement of SIRPα and PD-1, respectively, prevented phagocytosis by antigen-presenting cells and thereby limited TAA cross-presentation and innate immune activation. This postirradiation CD47 and PD-L1 up-regulation was observed across various human solid tumor cells. Concordantly, rectal cancer patients with poor responses to neoadjuvant RT exhibited significantly elevated postirradiation CD47 levels. The combination of RT, anti-SIRPα, and anti-PD-1 reversed adaptive immune resistance and drove efficient TAA cross-presentation, resulting in robust TAA-specific CD8 T cell priming, functional activation of T effectors, and increased T cell clonality and clonal diversity. We observed significantly higher complete response rates to RT/anti-SIRPα/anti-PD-1 in both irradiated and abscopal tumors and prolonged survival in three distinct murine CRC models, including a cecal orthotopic model. The efficacy of triple combination therapy was STING dependent as knockout animals lost most benefit of adding anti-SIRPα and anti-PD-1 to RT. Despite activation across the myeloid stroma, the enhanced dendritic cell function accounts for most improvements in CD8 T cell priming. These data suggest ATR-mediated CD47 and PD-L1 up-regulation as a key mechanism restraining radiation-induced immune priming. RT combined with SIRPα and PD-1 blockade promotes robust antitumor immune priming, leading to systemic tumor regressions.","prettyUrl":"hsieh-2022-si","following":false,"created":"02/24/2022","featured":false,"publishedDate":"02/24/2022","urlOrId":"hsieh-2022-si","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1d60516a-ac87-4d5b-8685-fb31d5aa2f5a","title":"Evidence of SARS-CoV-2-specific T-cell-mediated myocarditis in a MIS-A case","investigator":"Vannella, Kevin M.","investigatorInstitution":"Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health","publicationName":"Frontiers in Immunology","researchArea":"Other","prettyUrls":{"240":"vannella-2021-fi"},"prettyUrlList":["vannella-2021-fi"],"summary":"A 26-year-old otherwise healthy man died of fulminant myocarditis. Nasopharyngeal specimens collected premortem tested negative for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Histopathological evaluation of the heart showed myocardial necrosis surrounded by cytotoxic T-cells and tissue-repair macrophages. Myocardial T-cell receptor (TCR) sequencing revealed hyper-dominant clones with highly similar sequences to TCRs that are specific for SARS-CoV-2 epitopes. SARS-CoV-2 RNA was detected in the gut, supporting a diagnosis of multisystem inflammatory syndrome in adults (MIS-A). Molecular targets of MIS-associated inflammation are not known. Our data indicate that SARS-CoV-2 antigens selected high-frequency T-cell clones that mediated fatal myocarditis.","prettyUrl":"vannella-2021-fi","following":false,"created":"11/05/2021","featured":false,"publishedDate":"02/18/2022","urlOrId":"vannella-2021-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c6bbe2b1-a828-4df6-9c51-1dd37aff0525","title":"SARS-CoV-2 (COVID-19)-specific T-cell and B-cell responses in convalescent rheumatoid arthritis: monozygotic twins pair case observation","investigator":"Arruda, Lucas CM","investigatorInstitution":"Department of Clinical Science, Intervention and Technology, Karolinska Institute","publicationName":"Scandinavian Journal of Immunology","researchArea":"RA","prettyUrls":{"239":"arruda-2022-sji"},"prettyUrlList":["arruda-2022-sji"],"summary":"Rheumatoid arthritis (RA) patients present higher risk of SARS-CoV-2 infection (COVID-19), and proper management of the disease in this population requires a better understanding of how the immune system controls the virus. We analyzed the T cell and B cell phenotypes, and their repertoire in a pair of monozygotic twins with RA mismatched for COVID-19 infection. Twin- was not infected, while Twin+ was infected and effectively controlled the infection. We found no significant changes on the αβ T cell composition, while γδ T cells and B cells presented considerable expansion of memory population in Twin+ and robust T/B cell responses to several SARS-CoV-2 peptides. T cell receptor β/γ-chain and immunoglobulin heavy chain next-generation sequencing depicted a remarkable higher diversity in Twin+ compared with Twin-, despite no significant changes being found in variable/joining family usage. Repertoire overlap analyses showed that, although being identical twins, very few clones were shared between them, indicating that COVID-19 may lead to deep changes on the immune cell repertoire in RA patients. Altogether, our results indicate that RA patients may develop robust and persistent COVID-19-specific T/B cell responses; γδ T cells and B cells may play a key role in the management of COVID-19 in RA, and the infection may lead to a profound reshaping of immune cell receptor specificities.","prettyUrl":"arruda-2022-sji","following":false,"created":"02/15/2022","featured":false,"publishedDate":"02/15/2022","urlOrId":"arruda-2022-sji","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b3c01fd5-b111-4ec5-a4cb-88ad60618be8","title":"Tissue-specific features of the T cell repertoire after allogeneic hematopoietic cell transplantation in human and mouse","investigator":"DeWolf, Susan","investigatorInstitution":"Memorial Sloan Kettering Cancer Center","publicationName":"Science Translational Medicine","researchArea":"BMT / Stem Cell Transplant","prettyUrls":{"238":"dewolf-2022-gvhd"},"prettyUrlList":["dewolf-2022-gvhd"],"summary":"T cells are the central drivers of many inflammatory diseases, but the repertoire of tissue-resident T cells at sites of pathology in human organs remains poorly understood. We examined the site-specificity of T cell receptor (TCR) repertoires across tissues (5 to 18 tissues per patient) in prospectively collected autopsies of patients with and without graft-versus-host disease (GVHD), a potentially lethal tissue-targeting complication of allogeneic hematopoietic cell transplantation, and in mouse models of GVHD. Anatomic similarity between tissues was a key determinant of TCR repertoire composition within patients, independent of disease or transplant status. The T cells recovered from peripheral blood and spleens in patients and mice captured a limited portion of the TCR repertoire detected in tissues. Whereas few T cell clones were shared across patients, motif-based clustering revealed shared repertoire signatures across patients in a tissue-specific fashion. T cells at disease sites had a tissue-resident phenotype and were of donor origin based on single-cell chimerism analysis. These data demonstrate the complex composition of T cell populations that persist in human tissues at the end stage of an inflammatory disorder after lymphocyte-directed therapy. These findings also underscore the importance of studying T cell in tissues rather than blood for tissue-based pathologies and suggest the tissue-specific nature of both the endogenous and posttransplant T cell landscape.","prettyUrl":"dewolf-2022-gvhd","following":false,"created":"02/01/2022","featured":false,"publishedDate":"02/02/2022","urlOrId":"dewolf-2022-gvhd","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b81d545a-b051-4933-9a63-cc1a856663f6","title":"Primary germinal center-resident T follicular helper cells are a physiologically distinct subset of CXCR5hiPD-1hi T follicular helper cells","investigator":"Yeh, Chen-Hao","investigatorInstitution":"Department of Immunology","publicationName":"Immunity","researchArea":"Basic Immunology","prettyUrls":{"237":"yeh-2021-immunity"},"prettyUrlList":["yeh-2021-immunity"],"summary":"T follicular helper (Tfh) cells are defined by a Bcl6+CXCR5hiPD-1hi phenotype, but only a minor fraction of these reside in germinal centers (GCs). Here, we examined whether GC-resident and -nonresident Tfh cells share a common physiology and function. Fluorescently labeled, GC-resident Tfh cells in different mouse models were distinguished by low expression of CD90. CD90neg/lo GCTfh cells required antigen-specific, MHCII+ B cells to develop and stopped proliferating soon after differentiation. In contrast, nonresident, CD90hi Tfh (GCTfh-like) cells developed normally in the absence of MHCII+ B cells and proliferated continu- ously during primary responses. The TCR repertoires of both Tfh subsets overlapped initially but later diverged in association with dendritic cell-dependent proliferation of CD90hi GCTfh-like cells, suggestive of TCR-dependency seen also in TCR-transgenic adoptive transfer experiments. Furthermore, the transcriptomes of CD90neg/lo and CD90hi GCTfh-like cells were enriched in different functional pathways. Thus, GC- resident and nonresident Tfh cells have distinct developmental requirements and activities, implying distinct functions.","prettyUrl":"yeh-2021-immunity","following":false,"created":"01/25/2022","featured":false,"publishedDate":"01/25/2022","urlOrId":"yeh-2021-immunity","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"807cb88d-294a-4ce8-a39e-453ab391eea6","title":"DJ-1 depletion prevents immunoaging in T-cell compartments","investigator":"Hefeng, Feng","investigatorInstitution":"Department of Infection and Immunity","publicationName":"EMBO Reports","researchArea":"Immunosenescence","prettyUrls":{"236":"zeng-2022-embor"},"prettyUrlList":["zeng-2022-embor"],"summary":"Decline in immune function during aging increases susceptibility to different aging related diseases. However, the underlying molecular mechanisms, especially the genetic factors contributing to imbalance of naïve/memory T-cell subpopulations, still remain largely elusive. Here we show that loss of DJ-1 encoded by PARK7/DJ-1, causing early-onset familial Parkinson’s disease (PD), unexpectedly diminished signs of immunoaging in T-cell compartments of both human and mice. Compared with two gender-matched unaffected siblings of similar ages, the index PD patient with DJ-1 deficiency showed a decline in many critical immunoaging features, including almost doubled non-senescent T cells. The observation was further consolidated by the results in 45-week-old DJ-1 knockout mice. Our data demonstrated that DJ-1 regulates several immunoaging features via hematopoietic-intrinsic and naïve-CD8-intrinsic mechanisms. Mechanistically, DJ-1 depletion reduced oxidative phosphorylation (OXPHOS) and impaired TCR sensitivity in naïve CD8 T cells at a young age, accumulatively leading to a reduced aging process in T-cell compartments in older mice. Our finding suggests an unrecognized critical role of DJ-1 in regulating immunoaging, discovering a potent target to interfere with immunoaging- and aging-associated diseases.","prettyUrl":"zeng-2022-embor","following":false,"created":"01/05/2022","featured":false,"publishedDate":"01/17/2022","urlOrId":"zeng-2022-embor","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"978766f4-ea28-4961-a052-1469328d3073","title":"Expansion of Candidate HPV-specific T Cells in the Tumor Microenvironment During Chemoradiotherapy is Prognostic in HPV16+ Cancers","investigator":"Colbert, Lauren E.","investigatorInstitution":"Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center","publicationName":"Cancer Immunology Research","researchArea":"Cancer","prettyUrls":{"235":"colbert-2022-cir"},"prettyUrlList":["colbert-2022-cir"],"summary":"Human papillomavirus (HPV) infection causes 600,000 new cancers worldwide each year. HPV-related cancers express the oncogenic proteins E6 and E7, which could serve as tumor-specific antigens. It is not known whether immunity to E6 and E7 evolves during chemoradiotherapy or affects survival. Using T cells from 2 HPV16+ patients, we conducted functional T-cell assays to identify candidate HPV-specific T cells and common T-cell receptor motifs, which we then analyzed across 86 patients with HPV-related cancers. The HPV-specific clones and E7-related T-cell receptor motifs expanded in the tumor microenvironment over the course of treatment, whereas non-HPV-specific T cells did not. In HPV16+ patients, improved recurrence-free survival was associated with HPV-responsive T-cell expansion during chemoradiotherapy.","prettyUrl":"colbert-2022-cir","following":false,"created":"01/14/2022","featured":false,"publishedDate":"01/14/2022","urlOrId":"colbert-2022-cir","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a18e97e1-a48c-40ad-a350-5bffc127b25b","title":"Sensory nerves in melanoma impede the formation of tertiary lymphoid structures and anti-tumor immune responses","investigator":"Bunimovich, Yuri L.","investigatorInstitution":"Department of Dermatology, University of Pittsburgh","publicationName":"Cancer Immunology Research","researchArea":"Cancer","prettyUrls":{"234":"kruglov-2022-melanoma"},"prettyUrlList":["kruglov-2022-melanoma"],"summary":"Peripheral neurons comprise a critical component of the tumor microenvironment (TME). The role of the autonomic innervation in cancer has been firmly established. However, the effect of the afferent (sensory) neurons on tumor progression remains unclear. Utilizing surgical and chemical skin sensory denervation methods, we showed that afferent neurons supported the growth of melanoma tumors in vivo and demonstrated that sensory innervation limited the activation of effective anti-tumor immune responses. Specifically, sensory ablation led to improved leukocyte recruitment into tumors, with decreased presence of lymphoid and myeloid immunosuppressive cells and increased activation of T-effector cells within the TME. Cutaneous sensory nerves hindered maturation of intratumoral high endothelial venules (HEVs) and limited formation of mature tertiary lymphoid-like structures containing organized clusters of CD4+ T cells and B cells. Denervation further increased T-cell clonality and expanded the B-cell repertoire in the TME. Importantly, CD8a depletion prevented denervation-dependent anti-tumor effects. Finally, we observed that gene signatures of inflammation and the content of neuron-associated transcripts inversely correlated in human primary cutaneous melanomas, with the latter representing a negative prognostic marker of patient overall survival. Our results suggest that tumor-associated sensory neurons negatively regulate the development of protective anti-tumor immune responses within the TME, thereby defining a novel target for therapeutic intervention in the melanoma setting.","prettyUrl":"kruglov-2022-melanoma","following":false,"created":"01/06/2022","featured":false,"publishedDate":"01/06/2022","urlOrId":"kruglov-2022-melanoma","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"fdefd2d8-d001-4b30-85eb-82db9f89e7d4","title":"Multi-objective Optimization Reveals Time- and Dose-Dependent Inflammatory Cytokine-Mediated Regulation of Human Stem Cell Derived T-cell Development","investigator":"Edgar, John","investigatorInstitution":"University of British Columbia, School of Biomedical Engineering","publicationName":"NPJ Regenerative Medicine","researchArea":"Other","prettyUrls":{"233":"edgar-2021-npjrm"},"prettyUrlList":["edgar-2021-npjrm"],"summary":"The generation of T-cells from stem cells in vitro could provide an alternative source of cells for immunotherapies. T-cell development from hematopoietic stem and progenitor cells (HSPCs) is tightly regulated through Notch pathway activation by Delta-like (DL) ligands 1 and 4. Other molecules, such as stem cell factor (SCF) and interleukin (IL)-7, play a supportive role in regulating the survival, differentiation, and proliferation of developing T-cells. Numerous other signaling molecules influence T-lineage development in vivo, but little work has been done to understand and optimize their use for T-cell production. Using a defined engineered thymic niche system, we undertook a multi-stage statistical learning-based optimization campaign and identified IL-3 and tumor necrosis factor α (TNFα) as a stage- and dose-specific enhancers of cell proliferation and T-lineage differentiation. We used this information to construct an efficient three-stage process for generating conventional TCRαβ+CD8+ T-cells expressing a diverse TCR repertoire from blood stem cells. Our work provides new insight into T-cell development and a robust system for generating T-cells to enable clinical therapies for treating cancer and immune disorders.","prettyUrl":"edgar-2021-npjrm","following":false,"created":"11/05/2021","featured":false,"publishedDate":"01/05/2022","urlOrId":"edgar-2021-npjrm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1f95c069-5f08-40e2-9a5f-4adad63af53b","title":"Epstein-Barr Virus+ B Cells in Breast Cancer Immune Response: A Case Report","investigator":"Aran, Andrea","investigatorInstitution":"Immunology Unit, Institut de Biotecnologia i Biomedicina, and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona","publicationName":"Frontiers in Immunology","researchArea":"Cancer","prettyUrls":{"231":"aran-2021-fi"},"prettyUrlList":["aran-2021-fi"],"summary":"EBV-specific T cells have been recently described to be involved in fatal encephalitis and myocarditis in cancer patients after immune checkpoint therapies. Here, we report the study of a human triple-negative breast cancer tumor (TNBC) and EBV-transformed B cells obtained from a patient-derived xenograft (PDX) that progressed into a lymphocytic neoplasm named xenograft-associated B-cell lymphoma (XABCL). T-cell receptor (TCR) high-throughput sequencing was performed to monitor the T-cell clonotypes present in the different samples. Forty-three T-cell clonotypes were found infiltrating the XABCL tissue after three passes in mice along 6 months. Eighteen of these (42%) were also found in the TNBC biopsy. TCR infiltrating the XABCL tissue showed a very restricted T-cell repertoire as compared with the biopsy-infiltrating T cells. Consequently, T cells derived from the TNBC biopsy were expanded in the presence of the B-cell line obtained from the XABCL (XABCL-LCL), after which the TCR repertoire obtained was again very restricted, i.e., only certain clonotypes were selected by the B cells. A number of these TCRs had previously been reported as sequences involved in infection, cancer, and/or autoimmunity. We then analyzed the immunopeptidome from the XABCL-LCL, to identify putative B-cell-associated peptides that might have been expanding these T cells. The HLA class I and class II-associated peptides from XABCL-LCL were then compared with published repertoires from LCL of different HLA typing. Proteins from the antigen processing and presentation pathway remained significantly enriched in the XABCL-LCL repertoire. Interestingly, some class II-presented peptides were derived from cancer-related proteins. These results suggest that bystander tumor-infiltrating EBV+ B cells acting as APC may be able to interact with tumor-infiltrating T cells and influence the TCR repertoire in the tumor site.","prettyUrl":"aran-2021-fi","following":false,"created":"10/18/2021","featured":false,"publishedDate":"11/04/2021","urlOrId":"aran-2021-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"f8c69c15-19d2-46b2-99f2-951d276cd78b","title":"Response and recurrence correlates in individuals treated with neoadjuvant anti-PD-1 therapy for resectable oral cavity squamous cell carcinoma","investigator":"Liu, Sixue","investigatorInstitution":"Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles","publicationName":"Cell Reports Medicine","researchArea":"Cancer Immunotherapy","prettyUrls":{"217":"liu-2021-crm"},"prettyUrlList":["liu-2021-crm"],"summary":"Neoadjuvant PD-1 blockade may be efficacious in some individuals with high-risk, resectable oral cavity head and neck cancer. To explore correlates of response patterns to neoadjuvant nivolumab treatment and post-surgical recurrences, we analyzed longitudinal tumor and blood samples in a cohort of 12 individuals displaying 33% responsiveness. Pretreatment tumor-based detection of FLT4 mutations and PTEN signature enrichment favors response, and high tumor mutational burden improves recurrence-free survival. In contrast, preexisting and/or acquired mutations (in CDKN2A, YAP1, or JAK2) correlate with innate resistance and/or tumor recurrence. Immunologically, tumor response after therapy entails T cell receptor repertoire diversification in peripheral blood and intratumoral expansion of preexisting T cell clones. A high ratio of regulatory T to T helper 17 cells in pretreatment blood predicts low T cell receptor repertoire diversity in pretreatment blood, a low cytolytic T cell signature in pretreatment tumors, and innate resistance. Our study provides a molecular framework to advance neoadjuvant anti-PD-1 therapy for individuals with resectable head and neck cancer.","prettyUrl":"liu-2021-crm","following":false,"created":"08/19/2021","featured":false,"publishedDate":"11/02/2021","urlOrId":"liu-2021-crm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"5d98be71-6c39-4c8b-9e64-bf82d2edd9c7","title":"The Similarity of Class II HLA Genotypes Defines Patterns of Autoreactivity in Idiopathic Bone Marrow Failure Disorders","investigator":"Simona Pagliuca","investigatorInstitution":"Translational Hematology and Oncology Research Department, Cleveland Clinic","publicationName":"Blood","researchArea":"Autoimmune Disorders","prettyUrls":{"230":"pagliuca-2021-b"},"prettyUrlList":["pagliuca-2021-b"],"summary":"Idiopathic aplastic anemia (IAA) is a rare autoimmune bone marrow failure disorder initiated by a human leukocyte antigen (HLA)-restricted T cell response to unknown antigens. As for other autoimmune disorders, the predilection for certain HLA profiles seems to represent an etiologic factor, however, the structure-function patterns involved in the self-presentation in this disease remain unclear. Herein we analyzed the molecular landscape of HLA complexes of a cohort of 300 IAA patients and almost 3000 healthy and disease controls, by deeply dissecting their genotypic configurations, functional divergence, self-antigen binding capabilities and T cell receptor (TCR) repertoire specificities. Specifically, analysis of the evolutionary divergence of HLA genotypes (HED) showed that IAA patients carried class II HLA molecules whose antigen binding sites were characterized by a high level of structural homology, only partially explained by specific risk allele profiles. This pattern implies reduced HLA binding capabilities, confirmed by binding analysis of hematopoietic stem cell derived self-peptides. IAA phenotype was associated with the enrichment in a few amino acids at specific positions within the peptide-binding groove of DRB1 molecules, affecting the interface HLA-antigen-TCR β and potentially constituting the basis of T-cell dysfunction and autoreactivity. When analyzing associations with clinical outcomes, low HED was associated with risk of malignant progression and worse survival, underlying reduced tumor surveillance in clearing potential neoantigens derived from mechanisms of clonal hematopoiesis. Our data shed light on the immunogenetic risk associated with IAA etiology and clonal evolution, and on general pathophysiological mechanisms potentially involved also in other autoimmune disorders.","prettyUrl":"pagliuca-2021-b","following":false,"created":"10/29/2021","featured":false,"publishedDate":"10/29/2021","urlOrId":"pagliuca-2021-b","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"267e0152-3e1e-4f28-becd-ac2b83a31d19","title":"A Phase I, Open-Label, Dose-Escalation Study of the OX40 Agonist Ivuxolimab in Patients With Locally Advanced or Metastatic Cancers","investigator":"Diab, Adi","investigatorInstitution":"Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center","publicationName":"Clinical Cancer Research","researchArea":"Cancer Immunotherapy","prettyUrls":{"229":"diab-2021-ccr"},"prettyUrlList":["diab-2021-ccr"],"summary":"Purpose: Stimulation of effector T cells is an appealing immunotherapeutic approach in oncology. OX40 (CD134) is a co-stimulatory receptor expressed on activated CD4+ and CD8+ T cells. Induction of OX40 following antigen recognition results in enhanced T-cell activation, proliferation, and survival, and OX40 targeting shows therapeutic efficacy in preclinical studies. We report the monotherapy dose-escalation portion of a multicenter, phase I trial (NCT02315066) of ivuxolimab (PF-04518600), a fully human immunoglobulin G2 agonistic monoclonal antibody specific for human OX40.\n\nExperimental design: Adult patients (N = 52) with selected locally advanced or metastatic cancers received ivuxolimab 0.01-10 mg/kg. Primary endpoints were safety and tolerability. Secondary/exploratory endpoints included preliminary assessment of antitumor activity, and biomarker analyses.\n\nResults: The most common all-causality adverse events were fatigue (46.2%), nausea (28.8%), and decreased appetite (25.0%). Of 31 treatment-related adverse events, 30 (96.8%) were grade {less than or equal to}2. No dose-limiting toxicities occurred. Ivuxolimab exposure increased in a dose-proportionate manner from 0.3 to 10 mg/kg. Full peripheral blood target engagement occurred at {greater than or equal to}0.3 mg/kg. Three (5.8%) patients achieved a partial response, and disease control was achieved in 56% of patients. Increased CD4+ central memory T-cell proliferation and activation, and clonal expansion of CD4+ and CD8+ T cells in peripheral blood were observed at 0.1 to 3.0 mg/kg. Increased immune cell infiltrate and OX40 expression were evident in on-treatment tumor biopsies.\n\nConclusions: Ivuxolimab was generally well tolerated with on-target immune activation at clinically relevant doses, showed preliminary anti-tumor activity, and may serve as a partner for combination studies.\nExperimental Design: Adult patients (N = 52) with selected locally advanced or metastatic cancers received ivuxolimab 0.01–10 mg/kg. Primary endpoints were safety and tolerability. Secondary/exploratory endpoints included preliminary assessment of antitumor activity, and biomarker analyses.\n\nResults: The most common all-causality adverse events were fatigue (46.2%), nausea (28.8%), and decreased appetite (25.0%). Of 31 treatment-related adverse events, 30 (96.8%) were grade ≤2. No dose-limiting toxicities occurred. Ivuxolimab exposure increased in a dose-proportionate manner from 0.3 to 10 mg/kg. Full peripheral blood target engagement occurred at ≥0.3 mg/kg. Three (5.8%) patients achieved a partial response, and disease control was achieved in 56% of patients. Increased CD4+ central memory T-cell proliferation and activation, and clonal expansion of CD4+ and CD8+ T cells in peripheral blood were observed at 0.1 to 3.0 mg/kg. Increased immune cell infiltrate and OX40 expression were evident in on-treatment tumor biopsies.\n\nConclusions: Ivuxolimab was generally well tolerated with on-target immune activation at clinically relevant doses, showed preliminary anti-tumor activity, and may serve as a partner for combination studies.","prettyUrl":"diab-2021-ccr","following":false,"created":"08/04/2021","featured":false,"publishedDate":"10/25/2021","urlOrId":"diab-2021-ccr","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7d50ad87-88f0-4f00-b884-764c8d38ac10","title":"Positive and negative selection shape the human naïve B cell repertoire","investigator":"Meffre, Eric","investigatorInstitution":"Department of Immunobiology, Yale University","publicationName":"Journal of Clinical Investigation","researchArea":"Basic Immunology","prettyUrls":{"228":"chen-2021-jci"},"prettyUrlList":["chen-2021-jci"],"summary":"Although negative selection of developing B cells in the periphery is well described, yet poorly understood, evidence of naive B cell positive selection remains elusive. Using 2 humanized mouse models, we demonstrate that there was strong skewing of the expressed immunoglobulin repertoire upon transit into the peripheral naive B cell pool. This positive selection of expanded naive B cells in humanized mice resembled that observed in healthy human donors and was independent of autologous thymic tissue. In contrast, negative selection of autoreactive B cells required thymus-derived Tregs and MHC class II-restricted self-antigen presentation by B cells. Indeed, both defective MHC class II expression on B cells of patients with rare bare lymphocyte syndrome and prevention of self-antigen presentation via HLA-DM inhibition in humanized mice resulted in the production of autoreactive naive B cells. These latter observations suggest that Tregs repressed autoreactive naive B cells continuously produced by the bone marrow. Thus, a model emerged, in which both positive and negative selection shaped the human naive B cell repertoire and that each process was mediated by fundamentally different molecular and cellular mechanisms.","prettyUrl":"chen-2021-jci","following":false,"created":"10/25/2021","featured":false,"publishedDate":"10/25/2021","urlOrId":"chen-2021-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"5b364172-4316-4508-9d28-fcb3cd15e552","title":"Distinct tumor-infiltrating lymphocyte landscapes are associated with clinical outcomes in localized non-small cell lung cancer","investigator":"Federico, Lorenzo","investigatorInstitution":"Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center","publicationName":"Annals of Oncology","researchArea":"TIL","prettyUrls":{"227":"federico-2021-ao"},"prettyUrlList":["federico-2021-ao"],"summary":"Background: Despite the importance of tumor-infiltrating T lymphocytes (TILs) in cancer biology, the relationship between TIL phenotypes and their prognostic relevance for localized non-small cell lung cancer (NSCLC) has not been well established. \n\nPatients and Methods: Fresh tumor and normal adjacent tissue was prospectively collected from 150 patients with localized NSCLC. Tissue was comprehensively characterized by high-dimensional flow cytometry of TILs integrated with immunogenomic data from multiplex immunofluorescence, TCR sequencing, exome sequencing, RNA sequencing, targeted proteomics, and clinicopathologic features. \n\nResults: While neither the magnitude of TIL infiltration nor specific TIL subsets were significantly prognostic alone, the integration of high-dimensional flow cytometry data identified two major immunotypes (IM1 and IM2) that were predictive of recurrence-free survival independent of clinical characteristics. IM2 was associated with poor prognosis and characterized by the presence of proliferating TILs expressing CD103, PD-1, TIM3, and ICOS. Conversely, IM1 was associated with good prognosis and differentiated by an abundance of CD8+ T cells expressing cytolytic enzymes, CD4+ T cells lacking the expression of inhibitory receptors, and increased levels of B cell infiltrates and tertiary lymphoid structures. While increased B cell infiltration was associated with good prognosis, the best prognosis was observed in patients with tumors exhibiting high levels of both B cells and T cells. These findings were validated in patient tumors from The Cancer Genome Atlas. \n\nConclusions: Our study suggests that although the number of infiltrating T cells is not associated with patient survival, the nature of the infiltrating T cells, resolved in distinct TIL immunotypes, is prognostically relevant in NSCLC and may inform therapeutic approaches to clinical care.","prettyUrl":"federico-2021-ao","following":false,"created":"08/23/2021","featured":false,"publishedDate":"10/15/2021","urlOrId":"federico-2021-ao","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c1b97d54-c413-4565-8885-65f75dec11c8","title":"Cold and heterogeneous T cell repertoire is associated with copy number aberrations and loss of immune genes in small-cell lung cancer","investigator":"Chen, Ming","investigatorInstitution":"Zhejiang Cancer Hospital","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"226":"chen-2021-nc"},"prettyUrlList":["chen-2021-nc"],"summary":"Small-cell lung cancer (SCLC) is speculated to harbor complex genomic intratumor heterogeneity (ITH) associated with high recurrence rate and suboptimal response to immunotherapy. Here, using multi-region whole exome/T cell receptor (TCR) sequencing as well as immunohistochemistry, we reveal a rather homogeneous mutational landscape but extremely cold and heterogeneous TCR repertoire in limited-stage SCLC tumors (LS-SCLCs). Compared to localized non-small cell lung cancers, LS-SCLCs have similar predicted neoantigen burden and genomic ITH, but significantly colder and more heterogeneous TCR repertoire associated with higher chromosomal copy number aberration (CNA) burden. Furthermore, copy number loss of IFN-γ pathway genes is frequently observed and positively correlates with CNA burden. Higher mutational burden, higher T cell infiltration and positive PD-L1 expression are associated with longer overall survival (OS), while higher CNA burden is associated with shorter OS in patients with LS-SCLC.","prettyUrl":"chen-2021-nc","following":false,"created":"10/08/2021","featured":false,"publishedDate":"10/09/2021","urlOrId":"chen-2021-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d89ae3c0-685e-4051-b42f-02047df8ac08","title":"CD200 Blockade Modulates Tumor Immune Microenvironment but Fails to Show Efficacy in Inhibiting Tumor Growth in a Murine Model of Melanoma","investigator":"Talebian, Fatemeh","investigatorInstitution":"Department of Pathology, College of Medicine, The Ohio State University","publicationName":"Frontiers in Cell and Developmental Biology","researchArea":"Cancer","prettyUrls":{"221":"talebian-2021-fcdb"},"prettyUrlList":["talebian-2021-fcdb"],"summary":"CD200-CD200R pathway regulates immune responses and has been implicated in the pathogenesis of a number of cancer types. CD200 blockade is considered a strategy for immunotherapy of CD200-positive cancers such as melanoma. Thus, it is critical to understand the potential impacts of CD200 blockade in a more human relevant tumor model. In this study, we evaluated these issues using the CD200+ Yumm1.7 mouse melanoma model. Yumm1.7 cells bear Braf/Pten mutations resembling human melanoma. We found that Yumm1.7 tumors grow significantly faster in CD200R-/- mice compared to wild type mice. Analysis of tumor immune microenvironment (TIME) revealed that tumors from CD200R-/- or anti-CD200 treated mice had downregulated immune cell contents and reduced TCR clonality compared to tumors from untreated wild type mice. T cells also showed impaired effector functions, as reflected by reduced numbers of IFN-γ+ and TNF-α+ T cells. Mechanistically, we found upregulation of the CCL8 gene in CD200R-/- tumors. In vitro co-culture experiments using Yumm1.7 tumor cells with bone marrow derived macrophages (BMDM) from WT and CD200R-/- mice confirmed upregulation of macrophage CCL8 in the absence of CD200-CD200R interaction. Finally, we found that anti-CD200 therapy failed to show efficacy either alone or in combination with checkpoint inhibitors such as anti-PD-1 or anti-CTLA4 in inhibiting Yumm1.7 tumor growth. Given that CD200R-deficiency or anti-CD200 treatment leads to reduced T cell responses in TME, using blockade of CD200 as an immunotherapy for cancers such as melanoma should be practiced with caution.","prettyUrl":"talebian-2021-fcdb","following":false,"created":"09/02/2021","featured":false,"publishedDate":"09/20/2021","urlOrId":"talebian-2021-fcdb","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"2d1851db-cedd-4c68-9905-8ce5e406de02","title":"Proteogenomic Discovery of Neoantigens Facilitates Personalized Multi-antigen Targeted T cell Immunotherapy for Brain Tumors","investigator":"Rivero-Hinojosa, Samuel","investigatorInstitution":"Center for Cancer and Immunology Research, Children’s National Research Institute","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"224":"rivero-hinojosa-2021-nc"},"prettyUrlList":["rivero-hinojosa-2021-nc"],"summary":"Neoantigen discovery in pediatric brain tumors is hampered by their low mutational burden and scant tissue availability. We develop a low-input proteogenomic approach combining tumor DNA/RNA sequencing and mass spectrometry proteomics to identify tumor-restricted (neoantigen) peptides arising from multiple genomic aberrations to generate a highly target-specific, autologous, personalized T cell immunotherapy. Our data indicate that novel splice junctions are the primary source of neoantigens in medulloblastoma, a common pediatric brain tumor. Proteogenomically identified tumor-specific peptides are immunogenic and generate MHC II-based T cell responses. Moreover, polyclonal and polyfunctional T cells specific for tumor-specific peptides effectively eliminate tumor cells in vitro. Targeting novel tumor-specific antigens obviates the issue of central immune tolerance while potentially providing a safety margin favoring combination with other immune-activating therapies. These findings demonstrate the proteogenomic discovery of immunogenic tumor-specific peptides and lay the groundwork for personalized targeted T cell therapies for children with brain tumors.","prettyUrl":"rivero-hinojosa-2021-nc","following":false,"created":"09/17/2021","featured":false,"publishedDate":"09/20/2021","urlOrId":"rivero-hinojosa-2021-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1be67a99-528c-47fe-9fc9-30c58b9da621","title":"The AZD1222 COVID-19 vaccine induces a polyfunctional spike protein-specific Th1 response with a diverse TCR repertoire in humans","investigator":"Swanson II, Phillip A","investigatorInstitution":"Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health","publicationName":"Science Translational Medicine","researchArea":"Other","prettyUrls":{"223":"swanson-2021-stm"},"prettyUrlList":["swanson-2021-stm"],"summary":"AZD1222 (ChAdOx1 nCoV-19), a replication-deficient simian adenovirus-vectored vaccine, has demonstrated safety, efficacy, and immunogenicity against coronavirus disease 2019 (COVID-19) in clinical trials and real-world studies. We characterized CD4+ and CD8+ T cell responses induced by AZD1222 vaccination in peripheral blood mononuclear cells (PBMCs) from 296 unique vaccine recipients aged 18 to 85 years who enrolled in the phase 2/3 COV002 trial. Total spike protein-specific CD4+ T cell helper type 1 (Th1) and CD8+ T cell responses were increased in AZD1222-vaccinated adults of all ages following two doses of AZD1222. CD4+ Th2 responses following AZD1222 vaccination were not detected. Furthermore, AZD1222-specific Th1 and CD8+ T cells both displayed a high degree of polyfunctionality in all adult age groups. T cell receptor (TCR) β sequences from vaccinated participants mapped against TCR sequences known to react to SARS-CoV-2 revealed substantial breadth and depth across the SARS-CoV-2 spike protein for both AZD1222-induced CD4+ and CD8+ T cell responses. Overall, AZD1222 vaccination induced a polyfunctional Th1-dominated T cell response, with broad CD4+ and CD8+ T cell coverage across the SARS-CoV-2 spike protein.","prettyUrl":"swanson-2021-stm","following":false,"created":"08/16/2021","featured":false,"publishedDate":"09/14/2021","urlOrId":"swanson-2021-stm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"ec0fe735-586e-48ab-b646-9bcfbf44f139","title":"SASH3 variants cause a novel form of X-linked combined immunodeficiency with immune dysregulation","investigator":"Delmonte, Ottavia M.","investigatorInstitution":"Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy of Infectious Diseases, National Institutes of Health","publicationName":"Blood","researchArea":"Immunocompetence","prettyUrls":{"222":"delmonte-2021-b"},"prettyUrlList":["delmonte-2021-b"],"summary":"SAM and SH3 domain-containing 3 (SASH3), also called SH3-containing Lymphocyte Protein (SLY1) is a putative adaptor protein that is postulated to play an important role in the organization of signaling complexes and propagation of signal transduction cascades in lymphocytes. The SASH3 gene is located on the X-chromosome. Here, we identified three novel SASH3 deleterious variants in four unrelated male patients with a history of combined immunodeficiency and immune dysregulation manifesting as recurrent sinopulmonary, cutaneous and mucosal infections, and refractory autoimmune cytopenias. Patients exhibited CD4+ T cell lymphopenia, decreased T cell proliferation and cell cycle progression, and increased T cell apoptosis in response to mitogens. In vitro T-cell differentiation of CD34+ cells and molecular signatures of rearrangements at the T-cell receptor alpha (TRA) locus were indicative of impaired thymocyte survival. These patients also manifested neutropenia and B and NK cell lymphopenia. Lentivirus-mediated transfer of the SASH3 cDNA corrected protein expression, in vitro proliferation and signaling in SASH3-deficient Jurkat and patient-derived T cells. These findings define a new type of X-linked combined immunodeficiency in humans that recapitulates many of the abnormalities reported in Sly1-/- and Sly1D/Dmutant mice, highlighting an important role of SASH3 in human lymphocyte function and survival.","prettyUrl":"delmonte-2021-b","following":false,"created":"09/03/2021","featured":false,"publishedDate":"09/07/2021","urlOrId":"delmonte-2021-b","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"3028f52c-81cb-49dd-a146-4adf4389118d","title":"Next-Generation Sequencing-Based Monitoring of Circulating Tumor DNA Reveals Clonotypic Heterogeneity in Untreated PTCL","investigator":"Miljkovic, Milos D.","investigatorInstitution":"Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health","publicationName":"Blood Advances","researchArea":"Cancer","prettyUrls":{"220":"miljkovic-2021-ba"},"prettyUrlList":["miljkovic-2021-ba"],"summary":"Peripheral T-cell lymphomas (PTCL) have marked biologic and clinical heterogeneity, which confounds treatment decisions. Advances in circulating tumor DNA (ctDNA) assays employing next generation sequencing (NGS) has improved the detection of molecular relapse and driver mutations in diffuse large B-cell lymphoma, and highlight the potential utility of ctDNA across lymphomas. We investigated NGS-based monitoring of T-cell receptor (TCR) sequences in PTCL patients undergoing frontline treatment (NCT00001337). Of 45 patients, 34 (76%) had tumor-specific clonotypes of the TCR β or ɣ genes identified, which included 18 (86%) from baseline tissue and 16 (67%) from baseline serum. Twenty-five (74%) patients had both TCRβ and TCRɣ clonotypes, 23 (68%) patients had more than one TCRɣ clonotype, and 4 (9%) had multiple TCRβ or TCRɣ clonotypes, demonstrating significant intra-patient clonotypic heterogeneity. Among 24 patients with available serial serum samples during treatment, 9 (38%) cleared ctDNA after 2 cycles of therapy, and 11 (46%) patients had detectable ctDNA at the end of treatment. Patients with detectable ctDNA after therapy showed a trend towards worse survival. Notably, two patients with persistently detectable ctDNA after therapy remain in remission with 10-years of follow-up. Clonotypic heterogeneity in tumors and persistence despite long-term remission suggests variability in oncological potential.","prettyUrl":"miljkovic-2021-ba","following":false,"created":"09/01/2021","featured":false,"publishedDate":"09/02/2021","urlOrId":"miljkovic-2021-ba","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"bb683b7f-4f1f-4816-bfea-12e5b293e61f","title":"Humans with inherited T cell CD28 deficiency are susceptible to skin papillomaviruses but are otherwise healthy","investigator":"Casanova, Jean-Laurent","investigatorInstitution":"The Rockefeller University","publicationName":"Cell","researchArea":"Dermatology","prettyUrls":{"219":"beziat-2021-cell"},"prettyUrlList":["beziat-2021-cell"],"summary":"We study a patient with the human papilloma virus (HPV)-2-driven \"tree-man\" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.","prettyUrl":"beziat-2021-cell","following":false,"created":"09/01/2021","featured":false,"publishedDate":"09/02/2021","urlOrId":"beziat-2021-cell","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d1668756-c766-4721-a1db-a402dfccbe1b","title":"HSCT corrects primary immunodeficiency and immune dysregulation in patients with POMP-related autoinflammatory disease","investigator":"Poli, Cecilia","investigatorInstitution":"Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo","publicationName":"Blood","researchArea":"BMT / Stem Cell Transplant","prettyUrls":{"218":"martinez-2021-b"},"prettyUrlList":["martinez-2021-b"],"summary":"Inborn errors of immunity that present with concomitant immunodeficiency and auto-inflammation are therapeutically challenging; furthermore, complexity is added when they are caused by mutations in genes that encode for proteins expressed beyond immune cells. The ubiquitin-proteasome system is the main intracellular proteolytic machinery and participates in most cellular processes by degrading ubiquitinated proteins. Mutations in proteasome subunits resulting in proteasome deficiency cause a severe auto-inflammatory disease characterized by chronic auto-inflammation neutrophilic dermatosis and fever, collectively referred to as Proteasome Associated Auto-inflammatory Syndromes (PRAAS). POMP is a chaperone for proteasome assembly and AD mutations in POMP cause a form of PRAAS with prominent immunodeficiency referred to as POMP-related auto-inflammation and immune dysregulation (PRAID) manifesting with recurrent, severe and opportunistic infections in addition to inflammatory features that are characteristic for all PRAAS disorders, most importantly early-onset neutrophilic dermatosis. JAK inhibitors partially control the disease in individuals with PRAAS, however life-threatening, recurrent and opportunistic infections in patients with POMP mutations limit immunosuppressive therapies and prompted consideration of hematopoietic stem cell transplant (HSCT). We describe successful HSCT in two patients with POMP deficiency. Despite POMP being ubiquitously expressed, the immunologic and auto- inflammatory phenotype were both ameliorated through HSCT which suggests that the clinical and immunological features of PRAID are predominantly derived from a proteasome defect in hematopoietic cells. To our knowledge, these are the first patients with a form of PRAAS cured by HSCT, opening new therapeutic possibilities for these diseases.","prettyUrl":"martinez-2021-b","following":false,"created":"09/01/2021","featured":false,"publishedDate":"09/02/2021","urlOrId":"martinez-2021-b","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7bfb9e61-86b3-4569-87e4-0e6c04e04843","title":"Differential effects of PD-1 and CTLA-4 blockade on the melanoma-reactive CD8 T cell response","investigator":"Kvistborg, Pia","investigatorInstitution":"Netherlands Cancer Institute, Department of Molecular Oncology & Immunology","publicationName":"PNAS","researchArea":"Cancer Immunotherapy","prettyUrls":{"212":"gangaev-2021-pnas"},"prettyUrlList":["gangaev-2021-pnas"],"summary":"Immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have revolutionized the treatment of melanoma patients. Based on early studies addressing the mechanism of action, it was assumed that PD-1blockade mostly influences T cell responses at the tumor site. However, recent work has demonstrated that PD-1 blockade can influence the T cell compartment in peripheral blood. If activation of circulating tumor-reactive T cells would form an important mechanism of action ofPD-1 blockade, it may be predicted that such blockade would alter either the frequency and/or the breadth of the tumor-reactive CD8 T cell response. To address this question, we analyzed CD8 Tcell responses towards 71 melanoma associated epitopes in peripheral blood of 24 melanoma patients. We show that both the frequency and the breadth of the circulating melanoma-reactiveCD8 T cell response was unaltered upon PD-1 blockade. In contrast, a broadening of the circulating melanoma-reactive CD8 T cell response was observed upon CTLA-4 blockade, in concordance with our prior data. On the basis of these results, we conclude that PD-1 and CTLA-4 blockade have distinct mechanisms of action. In addition, the data provide an argument in favor of the hypothesis that anti-PD-1 therapy may primarily act at the tumor site.","prettyUrl":"gangaev-2021-pnas","following":false,"created":"07/16/2021","featured":false,"publishedDate":"08/20/2021","urlOrId":"gangaev-2021-pnas","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"2aecf67b-8f36-4389-85d2-2fb8b7e66d44","title":"Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial","investigator":"Scott J. Antonia","investigatorInstitution":"Duke Cancer Institute, Duke University School of Medicine","publicationName":"Nature Medicine","researchArea":"TIL","prettyUrls":{"216":"creelan-2021-nm"},"prettyUrlList":["creelan-2021-nm"],"summary":"Adoptive cell therapy using tumor-infiltrating lymphocytes (TILs) has shown activity in melanoma, but has not been previously evaluated in metastatic non-small cell lung cancer. We conducted a single-arm open-label phase 1 trial ( NCT03215810 ) of TILs administered with nivolumab in 20 patients with advanced non-small cell lung cancer following initial progression on nivolumab monotherapy. The primary end point was safety and secondary end points included objective response rate, duration of response and T cell persistence. Autologous TILs were expanded ex vivo from minced tumors cultured with interleukin-2. Patients received cyclophosphamide and fludarabine lymphodepletion, TIL infusion and interleukin-2, followed by maintenance nivolumab. The end point of safety was met according to the prespecified criteria of ≤17% rate of severe toxicity (95% confidence interval, 3-29%). Of 13 evaluable patients, 3 had confirmed responses and 11 had reduction in tumor burden, with a median best change of 35%. Two patients achieved complete responses that were ongoing 1.5 years later. In exploratory analyses, we found T cells recognizing multiple types of cancer mutations were detected after TIL treatment and were enriched in responding patients. Neoantigen-reactive T cell clonotypes increased and persisted in peripheral blood after treatment. Cell therapy with autologous TILs is generally safe and clinically active and may constitute a new treatment strategy in metastatic lung cancer.","prettyUrl":"creelan-2021-nm","following":false,"created":"08/17/2021","featured":false,"publishedDate":"08/19/2021","urlOrId":"creelan-2021-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"be54b338-f570-4477-afde-0756d9d3d27f","title":"The RAG1 N-terminal region regulates the efficiency and pathways of synapsis for V(D)J recombination","investigator":"Beilinison, Helen A","investigatorInstitution":"Department of Immunobiology, Yale School of Medicine, Yale University","publicationName":"Journal of Experimental Medicine","researchArea":"Basic Immunology","prettyUrls":{"215":"beilinson-2021-jem"},"prettyUrlList":["beilinson-2021-jem"],"summary":"Immunoglobulin and T cell receptor gene assembly depends on V(D)J recombination initiated by the RAG1-RAG2 recombinase. The RAG1 N-terminal region (NTR; aa 1-383) has been implicated in regulatory functions whose influence on V(D)J recombination and lymphocyte development in vivo are poorly understood. We generated mice in which RAG1 lacks ubiquitin ligase activity (P326G), the major site of autoubiquitination (K233R), or its first 215 residues (d215). While few abnormalities were detected in R1.K233R mice, R1.P326G mice exhibit multiple features indicative of reduced recombination efficiency including an increased Igk+:Igl+ B cell ratio and decreased recombination of Igh, Igk, Igl, and Tcrb loci. Previous studies indicate that synapsis of recombining partners during Igh recombination occurs through two pathways: long-range scanning and short-range collision. We find that R1d215 mice exhibit reduced short-range Igh and Tcrb D-to-J recombination. Our findings indicate that the RAG1 NTR regulates V(D)J recombination and lymphocyte development by multiple pathways including control of the balance between short- and long-range recombination.","prettyUrl":"beilinson-2021-jem","following":false,"created":"08/02/2021","featured":false,"publishedDate":"08/09/2021","urlOrId":"beilinson-2021-jem","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"322a46d5-7ed3-4749-9799-f0df1fe6d0a8","title":"Immunogenicity of Ad26.COV2.S vaccine against SARS-CoV-2 variants in humans","investigator":"Barouch, Dan H.","investigatorInstitution":"Harvard Medical School","publicationName":"Nature","researchArea":"Vaccine efficacy","prettyUrls":{"214":"alter-2021-n"},"prettyUrlList":["alter-2021-n"],"summary":"The Ad26.COV2.S vaccine1-3 has demonstrated clinical efficacy against symptomatic COVID-19, including against the B.1.351 variant that is partially resistant to neutralizing antibodies1. However, the immunogenicity of this vaccine in humans against SARS-CoV-2 variants of concern remains unclear. Here we report humoral and cellular immune responses from 20 Ad26.COV2.S vaccinated individuals from the COV1001 phase I-IIa clinical trial2 against the original SARS-CoV-2 strain WA1/2020 as well as against the B.1.1.7, CAL.20C, P.1 and B.1.351 variants of concern. Ad26.COV2.S induced median pseudovirus neutralizing antibody titres that were 5.0-fold and 3.3-fold lower against the B.1.351 and P.1 variants, respectively, as compared with WA1/2020 on day 71 after vaccination. Median binding antibody titres were 2.9-fold and 2.7-fold lower against the B.1.351 and P.1 variants, respectively, as compared with WA1/2020. Antibody-dependent cellular phagocytosis, complement deposition and natural killer cell activation responses were largely preserved against the B.1.351 variant. CD8 and CD4 T cell responses, including central and effector memory responses, were comparable among the WA1/2020, B.1.1.7, B.1.351, P.1 and CAL.20C variants. These data show that neutralizing antibody responses induced by Ad26.COV2.S were reduced against the B.1.351 and P.1 variants, but functional non-neutralizing antibody responses and T cell responses were largely preserved against SARS-CoV-2 variants. These findings have implications for vaccine protection against SARS-CoV-2 variants of concern.","prettyUrl":"alter-2021-n","following":false,"created":"07/27/2021","featured":false,"publishedDate":"07/28/2021","urlOrId":"alter-2021-n","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"9a376328-dfea-4679-8fb7-13e49580e4ff","title":"Evaluating T-cell cross-reactivity between tumors and immune-related adverse events with TCR sequencing: pitfalls in interpretations of functional relevance","investigator":"Smith, Kellie N","investigatorInstitution":"Department of Oncology, Johns Hopkins University School of Medicine","publicationName":"Journal for ImmunoTherapy of Cancer","researchArea":"Cancer","prettyUrls":{"213":"cottrell-2021-jitc"},"prettyUrlList":["cottrell-2021-jitc"],"summary":"T-cell receptor sequencing (TCRseq) enables tracking of T-cell clonotypes recognizing the same antigen over time and across biological compartments. TCRseq has been used to test if cross-reactive antitumor T cells are responsible for development of immune-related adverse events (irAEs) following immune checkpoint blockade. Prior studies have interpreted T-cell clones shared among the tumor and irAE as evidence supporting this, but interpretations of these findings are challenging, given the constraints of TCRseq. Here we capitalize on a rare opportunity to understand the impact of potential confounders, such as sample size, tissue compartment, and collection batch/timepoint, on the relative proportion of shared T-cell clones between an irAE and tumor specimens. TCRseq was performed on tumor-involved and -uninvolved tissues, including an irAE, that were obtained throughout disease progression and at the time of rapid autopsy from a patient with renal cell carcinoma treated with programmed death-1 (PD-1) blockade. Our analyses show significant effects of these confounders on our ability to understand T-cell receptor overlap, and we present mitigation strategies and study design recommendations to reduce these errors. Implementation of these strategies will enable more rigorous TCRseq-based studies of immune responses in human tissues, particularly as they relate to antitumor T-cell cross-reactivity in irAEs following checkpoint blockade.","prettyUrl":"cottrell-2021-jitc","following":false,"created":"07/20/2021","featured":false,"publishedDate":"07/21/2021","urlOrId":"cottrell-2021-jitc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"932112f9-dee2-4013-abb1-24d67aa3d74c","title":"Intrathecal activation of CD8+ memory T cells in IgG4-related disease of the brain parenchyma","investigator":"Friedrich, Mirco","investigatorInstitution":"German Cancer Research Center (DKFZ)","publicationName":"EMBO Molecular Medicine","researchArea":"Autoimmune Disorders","prettyUrls":{"211":"friedrich-2021-embomm"},"prettyUrlList":["friedrich-2021-embomm"],"summary":"IgG4-related disease (IgG4-RD) is a fibroinflammatory disorder signified by aberrant infiltration of IgG4-restricted plasma cells into a variety of organs. Clinical presentation is heterogeneous, and patho- physiological mechanisms of IgG4-RD remain elusive. There are very few cases of IgG4-RD with isolated central nervous system manifes- tation. By leveraging single-cell sequencing of the cerebrospinal fluid (CSF) of a patient with an inflammatory intracranial pseudotumor, we provide novel insights into the immunopathophysiology of IgG4- RD. Our data illustrate an IgG4-RD-associated polyclonal T-cell response in the CSF and an oligoclonal T-cell response in the parenchymal lesions, the latter being the result of a multifaceted cell–cell interaction between immune cell subsets and pathogenic B cells. We demonstrate that CD8+ T effector memory cells might drive and sustain autoimmunity via macrophage migration inhibitory factor (MIF)-CD74 signaling to immature B cells and CC-chemokine ligand 5 (CCL5)-mediated recruitment of cytotoxic CD4+ T cells. These findings highlight the central role of T cells in sustaining IgG4-RD and open novel avenues for targeted therapies.","prettyUrl":"friedrich-2021-embomm","following":false,"created":"05/04/2021","featured":false,"publishedDate":"07/07/2021","urlOrId":"friedrich-2021-embomm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"63b3fa7e-71ca-4260-8621-03d2cabc87d5","title":"Intrathymic differentiation of natural antibody-producing plasma cells in human neonates","investigator":"Cordero, Hector","investigatorInstitution":"Columbia University Medical Center","publicationName":"Nature Communications","researchArea":"Innate Immunity","prettyUrls":{"210":"cordero-2021-nc"},"prettyUrlList":["cordero-2021-nc"],"summary":"The thymus is a central lymphoid organ responsible for the development of T cells. Here, we show that the thymus of human neonates also contains a consistent contingent of CD138+ plasma cells, producing all classes and subclasses of immunoglobulins with the exception of IgD. These antibody-secreting cells (ASC) are comprised within a larger subset of B cells lacking expression of the complement receptors CD21 and CD35 and sharing the expression of signature genes defining mouse B1 cells. Single-cell transcriptomic, clonal correspondence and in vitro differentiation assays supported the intrathymic differentiation of CD138+ plasma cells alongside other B cell subsets with distinctive molecular phenotypes. Neonatal thymic plasma cells also included clones reactive to commensal and pathogenic bacteria that commonly infect children born with antibody deficiency. Thus, our findings point to the thymus as a source of innate humoral immunity in human neonates.","prettyUrl":"cordero-2021-nc","following":false,"created":"06/22/2021","featured":false,"publishedDate":"06/28/2021","urlOrId":"cordero-2021-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"f8dace30-0086-4b22-b1a7-6aeebdf24ede","title":"Clinical and basic implications of dynamic T cell receptor clonotyping in hematopoietic cell transplantation","investigator":"Simona Pagliuca","investigatorInstitution":"Translational Hematology and Oncology Research Program, Department of Hematology and Oncology, Cleveland Clinic Foundation","publicationName":"JCI insight","researchArea":"BMT / Stem Cell Transplant","prettyUrls":{"209":"pagliuca-2021-jcii"},"prettyUrlList":["pagliuca-2021-jcii"],"summary":"An effective T-cell receptor (TCR) repertoire diversification contributes to the success of hematopoietic cell transplantation (HCT). A full understanding of the patterns of T cell adaptive reconstitution may help in the discrimination of alloreactive responses. Herein, we prospectively studied the superstructure of TCR repertoire in 135 serial specimens from 35 patients receiving HCT for myeloid malignancies. \nPaired analysis of clonotypic repertoires showed a minimal overlap with donor expansions. Rarefied and hyperexpanded clonotypic patterns were the hallmarks of the T cell reconstitution and influenced clinical outcomes. Donor and pretransplant TCR diversity as well as divergence of class I human leukocyte antigen genotypes were major predictors of recipient TCR repertoire recovery. Complementary determining region 3–based specificity spectrum analysis indicated a predominant expansion of pathogen- and tumor-associated clonotypes in the late post–\nallo-HCT phase, while autoreactive clones were more expanded in case of graft-versus-host disease occurrence. These findings shed light on post–allo-HCT adaptive immune reconstitution processes and possibly help in tracking alloreactive responses.\n\n\n\nKeywords: TCR, adaptive immune competence, HCT, GVHD, clonal expansion\n\nKey points: \n\n•\tNGS deep TCR VβCDR3 sequencing allows for assessing clonal T-cell repertoire T-cell dynamics and tracing individual clonal specificities following hematopoietic stem cell transplantation.\n•\tAfter allogeneic hematopoietic cell transplantation, the restoration of TCR recognition spectrum is protracted and shaped by the post-transplant clinical course, diverging from the donor repertoire. \n•\tTCR VβCDR3 can serve as a quantifiable marker of specific immune responses defining a unique structure minimally overlapping with donor repertoire.","prettyUrl":"pagliuca-2021-jcii","following":false,"created":"06/24/2021","featured":false,"publishedDate":"06/28/2021","urlOrId":"pagliuca-2021-jcii","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"e146af14-2545-42e9-9a07-acf35b3521dc","title":"Low neoantigen expression and poor T cell priming underlie early immune escape in colorectal cancer","investigator":"Westcott, Peter MK","investigatorInstitution":"David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology","publicationName":"Nature Cancer","researchArea":"Cancer","prettyUrls":{"208":"westcott-2021-nc"},"prettyUrlList":["westcott-2021-nc"],"summary":"Immune evasion is a hallmark of cancer, and therapies that restore immune surveillance have proven highly effective in cancers with high tumor mutation burden (TMB) (e.g., those with microsatellite instability (MSI)). Whether low TMB cancers, which are largely refractory to immunotherapy, harbor potentially immunogenic neoantigens remains unclear. Here, we show that tumors from all patients with microsatellite stable (MSS) colorectal cancer (CRC) express clonal predicted neoantigens despite low TMB. Unexpectedly, these neoantigens are broadly expressed at lower levels compared to those in MSI CRC. Using a versatile platform for modulating neoantigen expression in CRC organoids and transplantation into the distal colon of mice, we show that low expression precludes productive cross priming and drives immediate T cell dysfunction. Strikingly, experimental or therapeutic rescue of priming rendered T cells capable of controlling tumors with low neoantigen expression. These findings underscore a critical role of neoantigen expression level in immune evasion and therapy response.","prettyUrl":"westcott-2021-nc","following":false,"created":"06/21/2021","featured":false,"publishedDate":"06/22/2021","urlOrId":"westcott-2021-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"dcecacac-f025-4057-894a-63e54a367790","title":"Expansion of Unique Hepatitis C Virus Specific Public CD8+ T Cell Clonotypes during Acute Infection and Reinfection","investigator":"Shoukry, Naglaa H","investigatorInstitution":"Département de médecine, Centre de Recherche du Centre Hospitalier de l’Université de Montréal","publicationName":"Journal of Immunology","researchArea":"Infectious Disease","prettyUrls":{"207":"mazouz-2021-ji"},"prettyUrlList":["mazouz-2021-ji"],"summary":"Hepatitis C virus (HCV) infection resolves spontaneously in approximately 25% of acutely infected humans where viral clearance is mediated primarily by virus specific CD8+ T cells. Previous cross-sectional analysis of the CD8+ T cell receptor (TCR) repertoire targeting two immunodominant HCV epitopes reported widespread use of public TCRs shared by different subjects, irrespective of infection outcome. However, little is known about the evolution of the public TCR repertoire during acute HCV and whether cross- reactivity to other antigens can influence infectious outcome. Here, we analyzed the CD8+ TCR repertoire specific to the immunodominant and cross-reactive HLA-A2 restricted NS3-1073 epitope during acute HCV in humans progressing to either spontaneous resolution or chronic infection, and at ~1-year following viral clearance. TCR repertoire diversity was comparable among all groups with preferential usage of the V04 and V06 gene families. We identified a set of thirteen public clonotypes in HCV-infected humans independent of infection outcome. Six public clonotypes used the V04 gene family. Several public clonotypes were long-lived in resolvers and expanded upon reinfection. By mining publicly available data, we identified several low frequency CDR3 sequences in the HCV-specific repertoire matching human TCRs specific for other HLA-A2 restricted epitopes from melanoma, cytomegalovirus, influenza A, Epstein-Barr and yellow fever viruses but they were of low frequency and limited cross-reactivity. In conclusion, we identified thirteen new public human CD8+ TCR clonotypes unique to HCV that expanded during acute infection and reinfection. The low frequency of cross- reactive TCRs suggest that they are not major determinants of infectious outcome.","prettyUrl":"mazouz-2021-ji","following":false,"created":"06/11/2021","featured":false,"publishedDate":"06/21/2021","urlOrId":"mazouz-2021-ji","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c598d17c-127f-4b3d-8a4e-47860f8b00c8","title":"IL-32 supports the survival of malignant T cells in cutaneous T cell lymphoma","investigator":"Clark, Rachael A.","investigatorInstitution":"Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School","publicationName":"Journal of Investigative Dermatology","researchArea":"Dermatology","prettyUrls":{"206":"yu-2021-jid"},"prettyUrlList":["yu-2021-jid"],"summary":"Cutaneous T-cell lymphoma (CTCL) is a heterogeneous collection of non-Hodgkin’s lymphomas derived from T cells that are tropic for the skin. Our understanding of this disease has been hampered by the fact that malignant T cells survive poorly in culture and do not survive well even in sophisticated xenograft models ( Townsend et al., 2016 ). Isolation and culture of T cells from CTCL skin lesions lead to an overgrowth of benign infiltrating T cells and loss of the malignant T-cell clone. It is an inescapable irony that malignant T cells capable of killing patients die in even the most supportive in vitro and in vivo environments. These cells appear to require a survival factor found in the skin but not replicated by in vitro and in vivo murine systems.","prettyUrl":"yu-2021-jid","following":false,"created":"06/03/2021","featured":false,"publishedDate":"06/03/2021","urlOrId":"yu-2021-jid","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"08e34f68-0817-4493-a12f-088cb3aa6b88","title":"Transcriptional programs of neoantigen- specific TIL in anti-PD-1-treated lung cancers","investigator":"Smith, Kellie","investigatorInstitution":"Johns Hopkins Unversity","publicationName":"Nature","researchArea":"Cancer","prettyUrls":{"205":"caushi-2021-n"},"prettyUrlList":["caushi-2021-n"],"summary":"PD-1 blockade unleashes CD8 T cells, including those specific for mutation-associated neoantigens (MANA), but factors in the tumour microenvironment can inhibit these T cell responses. Single-cell transcriptomics have revealed global T cell dysfunctionprograms in tumour-infltrating lymphocytes (TIL). However, the majority of TIL do not recognize tumour antigens, and little is known about transcriptional programs of MANA-specific TIL. Here, we identify MANA-specific T cell clones using the MANA functional expansion of specific T cells assay in neoadjuvant anti-PD-1-treated non-small cell lung cancers (NSCLC). We use their T cell receptors as a ‘barcode’ to track and analyse their transcriptional programs in the tumour microenvironment using coupled single-cell RNA sequencing and T cell receptor sequencing. We found both MANA- and virus-specific clones in TIL, regardless of response, and MANA-, influenza- and Epstein–Barr virus-specific TIL each have unique transcriptional programs. Despite exposure to cognate antigen, MANA-specific TILs express an incompletely activated cytolytic program. MANA-specific CD8 T cells have hallmark transcriptional programs of tissue-resident memory (TRM) cells, but low levels of interleukin-7 receptor (IL-7R) and are functionally less responsive to interleukin-7 (IL7) compared with influenza-specific TRM cells. Compared with those from responding tumours, MANA-specific clones from non-responding tumours express T cell receptors with markedly lower ligand-dependent signaling, are largely confined to HOBIThigh TRM subsets, and coordinately upregulate checkpoints, killer inhibitory receptors and inhibitors of T cell activation. These findings provide important insights for overcoming resistance to PD-1 blockade.","prettyUrl":"caushi-2021-n","following":false,"created":"04/19/2021","featured":false,"publishedDate":"06/02/2021","urlOrId":"caushi-2021-n","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7b39cc48-f684-4c97-8a08-aa5386cebb27","title":"Single cell analysis of host response to helminth infection reveals the clonal breadth, heterogeneity, and tissue-specific programming of the responding CD4+ T cell repertoire","investigator":"Reinhardt, Richard Lee","investigatorInstitution":"National Jewish Health","publicationName":"PLoS Pathogens","researchArea":"Infectious Disease","prettyUrls":{"204":"brown-20201-pp"},"prettyUrlList":["brown-20201-pp"],"summary":"The CD4+ T cell response is critical to host protection against helminth infection. How this response varies across different hosts and tissues remains an important gap in our understanding. Using IL-4-reporter mice to identify responding CD4+ T cells to Nippostrongylus brasiliensis infection, T cell receptor sequencing paired with novel clustering algorithms revealed a broadly reactive and clonally diverse CD4+ T cell response. While the most prevalent clones and clonotypes exhibited some tissue selectivity, most were observed to reside in both the lung and lung-draining lymph nodes. Antigen-reactivity of the broader repertoires was predicted to be shared across both tissues and individual mice. Transcriptome, trajectory, and chromatin accessibility analysis of lung and lymph-node repertoires revealed three unique but related populations of responding IL-4+ CD4+ T cells consistent with T follicular helper, T helper 2, and a transitional population sharing similarity with both populations. The shared antigen reactivity of lymph node and lung repertoires combined with the adoption of tissue-specific gene programs allows for the pairing of cellular and humoral responses critical to the orchestration of anti-helminth immunity.","prettyUrl":"brown-20201-pp","following":false,"created":"02/10/2021","featured":false,"publishedDate":"05/18/2021","urlOrId":"brown-20201-pp","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"5609bfe7-453d-4777-8a2d-515c45ce6756","title":"Clonal analysis of immunodominance and cross-reactivity of the CD4 T cell response to SARS-CoV-2","investigator":"Cassotta, Antonino","investigatorInstitution":"Institute for Research in Biomedicine","publicationName":"Science","researchArea":"Infectious Disease","prettyUrls":{"203":"low-2021-s"},"prettyUrlList":["low-2021-s"],"summary":"The identification of CD4+ T cell epitopes is instrumental for the design of subunit vaccines for broad protection against coronaviruses. Here we demonstrate in COVID-19-recovered individuals a robust CD4+ T cell response to naturally processed SARS-CoV-2 spike (S) and nucleoprotein (N), including effector, helper, and memory T cells. By characterizing 2943 S-reactive T cell clones from 34 individuals, we found that RBD is highly immunogenic, and that 33% of RBD-reactive clones and 94% of individuals recognized a conserved immunodominant S346-365 region comprising nested HLA-DR- and HLA-DP-restricted epitopes. Using pre- and post-COVID-19 samples and S proteins from endemic coronaviruses, we identify cross-reactive T cells targeting multiple S protein sites. The immunodominant and cross-reactive epitopes identified can inform vaccination strategies to counteract emerging SARS-CoV-2 variants.","prettyUrl":"low-2021-s","following":false,"created":"04/14/2021","featured":false,"publishedDate":"05/14/2021","urlOrId":"low-2021-s","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"e74ca34e-3767-4e93-8060-a4711a0adb9c","title":"Distinct populations of antigen specific tissue resident CD8 T cells in human cervix mucosa","investigator":"Tao Peng","investigatorInstitution":"University of Washington","publicationName":"JCI Insight","researchArea":"Other","prettyUrls":{"202":"peng-2021-jcii"},"prettyUrlList":["peng-2021-jcii"],"summary":"The ectocervix is part of the lower female reproductive tract (FRT), which is susceptible to sexually transmitted infections (STI). Comprehensive knowledge of the phenotypes and T cell receptor (TCR) repertoire of tissue resident memory T cells (TRM) in human FRT is lacking. We have taken single-cell RNA sequencing approaches to simultaneously define gene expression and TCR clonotypes of the human ectocervix. There are significantly more CD8 than CD4 T cells. Unsupervised clustering and trajectory analysis identify distinct populations of CD8 T cells with IFNGhiGZMBlowCD69hiCD103low or IFNGlowGZMBhiCD69medCD103hi phenotypes. Little overlap was seen between their TCR repertoires. Immunofluorescent staining shows that CD103+ CD8 TRM cells preferentially localize in epithelium while CD69+ CD8 TRM distribute evenly in epithelium and stroma. Ex vivo assays indicate up to 14% of cervical CD8 TRM clonotypes are HSV-2 reactive in HSV-2-seropositive persons, reflecting physiologically relevant localization. Our studies identify subgroups of CD8 TRM in the human ectocervix that exhibit distinct expression of antiviral defense and tissue residency markers, anatomic locations, and TCR repertoires that target anatomically relevant viral antigens. Optimization of the location, number, and function of FRT TRM is an important approach for improving host defenses to STI.","prettyUrl":"peng-2021-jcii","following":false,"created":"05/10/2021","featured":false,"publishedDate":"05/10/2021","urlOrId":"peng-2021-jcii","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1e3ed1e4-b333-4989-9fab-fb9554cdd30e","title":"Functional characterization of CD4+ T-cell receptors cross-reactive for SARS-CoV-2 and endemic coronaviruses","investigator":"Smith, Kellie","investigatorInstitution":"Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Medicine","publicationName":"Journal of Clinical Investigation","researchArea":"Infectious Disease","prettyUrls":{"200":"dykema-2021-jci"},"prettyUrlList":["dykema-2021-jci"],"summary":"Background: Recent studies have reported T cell immunity to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in unexposed donors, possibly due to cross-recognition by T-cells specific for common cold coronaviruses (CCCs). True T-cell cross-reactivity, defined as the recognition by a single TCR of more than one distinct peptide-MHC ligand, has never been shown in the context of SARS-CoV-2.\n\nMethods: We used the ViraFEST platform to identify T cell responses cross-reactive for the spike (S) glycoproteins of SARS-CoV-2 and CCCs at the T cell receptor (TCR) clonotype level in convalescent COVID-19 patients (CCPs) and SARS-CoV-2-unexposed donors. Confirmation of SARS-CoV-2/CCC cross-reactivity and assessments of functional avidity were performed using a TCR cloning and transfection system.\n\nResults: Memory CD4+ T-cell clonotypes that cross-recognized the S proteins of SARS-CoV-2 and at least one other CCC were detected in 65% of CCPs and unexposed donors. Several of these TCRs were shared among multiple donors. Cross-reactive T-cells demonstrated significantly impaired SARS-CoV-2-specific proliferation in vitro relative to mono-specific CD4+ T-cells, which was consistent with lower functional avidity of their TCRs for SARS CoV-2 relative to CCC.\n\nConclusions: For the first time, our data confirm the existence of unique memory CD4+ T cell clonotypes cross-recognizing SARS-CoV-2 and CCCs. The lower avidity of cross-reactive TCRs for SARS-CoV-2 may be the result of antigenic imprinting, such that pre-existing CCC-specific memory T cells have reduced expansive capacity upon SARS-CoV-2 infection. Further studies are needed to determine how these cross-reactive T-cell responses impact clinical outcomes in COVID-19 patients.","prettyUrl":"dykema-2021-jci","following":false,"created":"04/21/2021","featured":false,"publishedDate":"04/22/2021","urlOrId":"dykema-2021-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"6642558a-918a-490e-bb39-78c12fe3a733","title":"Select antitumor cytotoxic CD8+ T clonotypes expand in patients with chronic lymphocytic leukemia treated with ibrutinib","investigator":"Baptista, Maria Joao","investigatorInstitution":"Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health","publicationName":"Clinical Cancer Research","researchArea":"Response to Therapeutic Agent","prettyUrls":{"199":"baptista-2021-ccr"},"prettyUrlList":["baptista-2021-ccr"],"summary":"Purpose: In chronic lymphocytic leukemia (CLL), the T-cell receptor (TCR) repertoire is skewed and tumor-derived antigens are hypothesized as drivers of oligoclonal expansion. Ibrutinib, a standard treatment for CLL, inhibits not only Bruton tyrosine kinase of the B-cell receptor signaling pathway, but also interleukin-2-inducible kinase of the TCR signaling pathway. T-cell polarization and activation are affected by ibrutinib, but it is unknown if T cells contribute to clinical response.\n\nExperimental Design: High-throughput TCRß sequencing was performed in 77 longitudinal samples from 26 CLL patients treated with ibrutinib. TCRß usage in CD4+ and CD8+ T cells and granzyme B expression were assessed by flow cytometric analysis. Antitumor cytotoxicity of T cells expanded with autologous CLL cells or with antigen-independent anti-CD3/CD28/CD137 beads was tested.\n\nResults: The clonality of the TCR repertoire increased at the time of response. With extended treatment, TCR clonality remained stable in patients with sustained remission and decreased in patients with disease progression. Expanded clonotypes were rarely shared between patients, indicating specificity for private antigens. Flow cytometry demonstrated a predominance of CD8+ cells among expanded clonotypes. Importantly, bulk T cells from responding patients were cytotoxic against autologous CLL cells in vitro and selective depletion of major expanded clonotypes reduced CLL cell killing.\n\nConclusions: In patients with CLL, established T-cell responses directed against tumor are suppressed by disease and reactivated by ibrutinib.","prettyUrl":"baptista-2021-ccr","following":false,"created":"02/23/2021","featured":false,"publishedDate":"04/20/2021","urlOrId":"baptista-2021-ccr","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"127959a5-a7c9-4477-9f2e-06705f9f8ba0","title":"B cell signatures and tertiary lymphoid structures contribute to outcome in head and neck squamous cell carcinoma","investigator":"Bruno, Tullia","investigatorInstitution":"University of Pittsburgh, Department of Immunology","publicationName":"Nature Communications","researchArea":"Cancer Immunotherapy","prettyUrls":{"198":"ruffin-2021-nc"},"prettyUrlList":["ruffin-2021-nc"],"summary":"Current immunotherapy paradigms aim to reinvigorate CD8+ T cells, but the contribution of humoral immunity to antitumor immunity remains understudied. Here, we demonstrate that in head and neck squamous cell carcinoma (HNSCC) caused by human papillomavirus infection (HPV+), patients have transcriptional signatures of germinal center (GC) tumor infiltrating B cells (TIL-Bs) and spatial organization of immune cells consistent with tertiary lymphoid structures (TLS) with GCs, both of which correlate with favorable outcome. GC TIL-Bs in HPV+ HNSCC are characterized by distinct waves of gene expression consistent with dark zone, light zone and a transitional state of GC B cells. Semaphorin 4a (SEMA4A) expression is enhanced on GC TIL-Bs present in TLS of HPV+ HNSCC and during the differentiation of TIL-Bs. Our study suggests that novel therapeutics to enhance TIL-B responses in HNSCC should be prioritized in future studies to determine if they can complement current T cell mediated immunotherapies.","prettyUrl":"ruffin-2021-nc","following":false,"created":"04/12/2021","featured":false,"publishedDate":"04/15/2021","urlOrId":"ruffin-2021-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1d4f576f-39a9-44e7-8540-4c9bf457f51f","title":"Generation of self-reactive, shared T-cell receptor α chains in the human thymus","investigator":"Arstila, T.Petteri","investigatorInstitution":"Research Programs Unit, Translational Immunology, and Medicum, University of Helsinki","publicationName":"Journal of Autoimmunity","researchArea":"Basic Immunology","prettyUrls":{"197":"heikkila-2021-ja"},"prettyUrlList":["heikkila-2021-ja"],"summary":"T-cell receptors (TCRs) are heterodimers consisting of TCRa and TCRb chains that are generated in a semistochastic process of genetic recombination. The estimated total diversity exceeds 10^8 unique TCRs. We have analyzed TCR clonotypes at their early developmental stage in thymus showed earlier that a surprisingly high fraction of TCR clonotypes are shared between unrelated individuals, concerning particularly TCRa chains with abundant clone sizes. Here, we studied the generation of TCRs associated with antigens that are previously identified in the context of type 1 diabetes (T1D) and HIV. We show that the human thymus prefers the generation of T1D-associated TCRa chains over HIV-associated chains. Furthermore, analysis of circulating repertoires shows that these T1D-associated chains are not lost in the negative selection nor predominantly converted to regulatory T-cell lineage.","prettyUrl":"heikkila-2021-ja","following":false,"created":"04/07/2021","featured":false,"publishedDate":"04/07/2021","urlOrId":"heikkila-2021-ja","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"cd09abd3-5c8b-465f-ae98-bf3f810b3229","title":"Treatment-induced immune cell priming as a potential explanation for an outstanding anti-tumor response in a patient with metastatic colorectal cancer","investigator":"Duhen, Thomas","investigatorInstitution":"Earle A. Chiles Research Institute | Providence Cancer Institute","publicationName":"Cancer Immunology Research","researchArea":"Cancer Immunotherapy","prettyUrls":{"196":"rajamanickam-2021-cir"},"prettyUrlList":["rajamanickam-2021-cir"],"summary":"Microsatellite stable (MSS) colorectal cancers (CRC) are known to harbor low mutation burden, a limited immune cell infiltration and thereby respond poorly to immunotherapy. Here we report a case of metastatic CRC with an outstanding anti-cancer immune response. Briefly, the primary tumor was resected in 2012 and the patient received several cycles of chemotherapy until 2017. In 2018, he underwent a left hepatectomy to remove a new metastasis. Analysis of this tumor revealed a strong CD8 T cell immune response. A high frequency of CD8 T cells co-expressed CD39 and CD103, a phenotype characteristic of tumor-reactive cells. Using whole exome sequencing, we identified somatic mutations recognized by CD39+CD103+ CD8 T cells. The observed reactivity against the tumor was dominated by the response to a single mutation that emerged in the colorectal liver metastasis. Importantly, somatic mutations that were not immunogenic in the primary tumor led to robust CD8 T cell expansion later during disease progression. Our data suggest that the cytotoxic treatment regimen received by the patient might be responsible for this effect. Hence, the capacity of cytotoxic regimens to prime the immune system in CRC patients should be investigated further and might provide a rationale for combination with immunotherapy.","prettyUrl":"rajamanickam-2021-cir","following":false,"created":"04/02/2021","featured":false,"publishedDate":"04/05/2021","urlOrId":"rajamanickam-2021-cir","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"062d2746-499b-4052-8799-5af771f655df","title":"Thymic atrophy creates holes in Treg-mediated immuno-regulation via impairment of an antigen-specific clone","investigator":"Su, Dong-Ming","investigatorInstitution":"Dept. of Microbiology, Immunology and Genetics, University of North Texas Health Science Center","publicationName":"Immunology","researchArea":"Basic Immunology","prettyUrls":{"195":"thomas-2021-immunology"},"prettyUrlList":["thomas-2021-immunology"],"summary":"Age-related thymic involution results in reduced output of naïve conventional T (Tcon) cells. However, its impact on regulatory T (Treg) cells is insufficiently understood. Given evidence that thymic Treg (tTreg) cell generation is enhanced in the aged, involuted thymus and that the aged periphery accumulates peripheral Treg (pTreg) cells, we asked why these Treg cells are unable to effectively attenuate increased auto-reactivity induced chronic inflammation in the elderly. We utilized a mock self-antigen chimeric mouse model, in which membrane-bound ovalbumin (mOVA) transgenic mice bearing a FoxN1-floxed gene for induction of conditional thymic involution, received MHC-II restricted, OVA-recognizing T cell receptor (TCR) transgenic progenitor cells. The chimeric mice with thymic involution exhibited a significant decrease in OVA-specific tTreg and pTreg cells, but not polyclonal (pan)-Treg cells. The OVA-specific pTreg cells were significantly less able to suppress OVA stimulation induced proliferation in vitro. Further, these cells exhibited intrinsic defects, displaying lower IL-2R expression coupled with lower FoxP3 expression upon OVA stimulation. Additionally, we conducted preliminary TCR repertoire diversity sequencing for Treg cells among recent thymic emigrants (RTEs) from RagGFP-FoxP3RFP dual reporter mice and observed a trend for decreased diversity in mice with thymic involution compared to littermates with normal thymus. In sum, these data indicate that although the effects of age-related thymic involution do not affect pan-Treg generation, a certain tissue-specific Treg clone is intrinsically impaired and not undergoing normal agonist selection, which could result in inability to attenuate age-related chronic inflammation.","prettyUrl":"thomas-2021-immunology","following":false,"created":"03/22/2021","featured":false,"publishedDate":"03/24/2021","urlOrId":"thomas-2021-immunology","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"f8c5743d-ff44-47fc-88a7-0609e801eed6","title":"Complete Response to Programmed Cell Death-1 Blockade Following EBV-specific Adoptive T-cell Therapy in a Patient with Metastatic Nasopharyngeal Carcinoma","investigator":"Corey Smith","investigatorInstitution":"QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute","publicationName":"NJP Precision Oncology","researchArea":"Adoptive T Cell Therapy","prettyUrls":{"194":"smith-2021-njppo"},"prettyUrlList":["smith-2021-njppo"],"summary":"Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV) associated heterogeneous disease and is characterized by peri-tumoral immune infiltrate. Adoptive T-cell therapy (ACT) has emerged as a potential therapeutic strategy for NPC. However, the tumour microenvironment remains a major roadblock for successful implementation of ACT in clinical settings. Expression of checkpoint molecules by malignant cells can inhibit the effector function of adoptively transferred EBV-specific T cells. Here we present a novel case report of a patient with metastatic NPC who was successfully treated with a combination of EBV-specific ACT and programmed cell death-1 blockade therapy. Following combination immunotherapy, the patient showed complete resolution of metastatic disease with no evidence of disease relapse for 22 months. Follow up immunological analysis revealed dramatic restructuring of the global T-cell repertoire that was coincident with the clinical response. This case report provides an important platform for translating these findings to a larger cohort of NPC patients.","prettyUrl":"smith-2021-njppo","following":false,"created":"01/31/2021","featured":false,"publishedDate":"03/22/2021","urlOrId":"smith-2021-njppo","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7577484f-af76-4f3d-b651-7ca4cfa6159f","title":"Clonally focused public and private T cells in resected brain tissue from surgeries to treat children with intractable seizures.","investigator":"Owens, Geoffrey C.","investigatorInstitution":"Department of Neurosurgery, David Geffen School of Medicine at UCLA","publicationName":"Frontiers in Immunology","researchArea":"Other","prettyUrls":{"193":"chang-2021-fi"},"prettyUrlList":["chang-2021-fi"],"summary":"Using a targeted transcriptomics approach, we have analyzed resected brain tissue from a cohort of 53 pediatric epilepsy surgery cases, and have found that there is a spectrum of involvement of both the innate and adaptive immune systems as evidenced by the differential expression of immune-specific genes in the affected brain tissue. The specimens with the highest expression of immune-specific genes were from two Rasmussen encephalitis cases, which is known to be a neuro-immunological disease, but also from tuberous sclerosis complex (TSC), focal cortical dysplasia, and hemimegalencephaly surgery cases. We obtained T cell receptor (TCR) V chain sequence data from brain tissue and blood from patients with the highest levels of T cell transcripts. The clonality indices and the frequency of the top 50 clonotypes indicated that T cells in the brain were clonally restricted. The top 50 clonotypes comprised both public and private (patient specific) clonotypes, and the TCR V chain third complementarity region (CDR3) of the most abundant public clonotype in each brain sample was strikingly similar to a CDR3 that recognizes an immunodominant epitope in either human cytomegalovirus or Epstein Barr virus, or influenza virus A. We found that the frequency of 14 of the top 50 brain clonotypes from a TSC surgery case had significantly increased in brain tissue removed to control recurrent seizures 11 months after the first surgery. Conversely, we found that the frequency in the blood of 18 of the top 50 brain clonotypes from a second TSC patient, who was seizure free, had significantly decreased 5 months after surgery indicating that T cell clones found in the brain had contracted in the periphery after removal of the brain area associated with seizure activity and inflammation. However, the frequency of a public and a private clonotype significantly increased in the brain after seizures recurred and the patient underwent a second surgery. Combined single cell gene expression and TCR sequencing of brain-infiltrating leukocytes from the second surgery showed that the two clonotypes were CD8 effector T cells, indicating that they are likely to be pathologically relevant.","prettyUrl":"chang-2021-fi","following":false,"created":"03/16/2021","featured":false,"publishedDate":"03/17/2021","urlOrId":"chang-2021-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"95fadbc3-34c7-4475-85a6-b06fb0c58a48","title":"Response to Hypomethylating Agents in Myelodysplastic Syndrome Is Associated with Emergence of Novel TCR Clonotypes","investigator":"Abbas, Hussein A.","investigatorInstitution":"University of Texas MD Anderson Cancer Center","publicationName":"Frontiers in Immunology","researchArea":"Response to Therapeutic Agent","prettyUrls":{"192":"abbas-2021-fi"},"prettyUrlList":["abbas-2021-fi"],"summary":"Aberrant T-cell function is implicated in the pathogenesis of myelodysplastic syndrome (MDS). Monitoring the T-cell receptor (TCR) repertoire can provide insights into T-cell adaptive immunity. Previous studies found skewed TCR repertoires in MDS compared to healthy patients, however these studies used mRNA-based spectratyping have limitations. Furthermore, evaluating the TCR repertoire in context of hypomethylating agents (HMAs) treatment can provide insights into the dynamics of T-cell mediated responses in MDS. We conducted immunosequencing of the CDR3 regions of TCRβ chains in bone marrows of 11 MDS patients prior (n=11) and following treatment (n=26) with hypomethylating agents, alongside bone marrows from 4 healthy donors as controls. TCR repertoires in MDS patients were more clonal and less diverse than healthy donors. However, unlike previous reports, we did not observe significant skewness in CDR3 length or spectratyping. The global metrics of TCR profiling including richness, clonality, overlaps were not significantly changed was observed in MDS patients following treatment with HMAs regardless of response. However, an emergence of novel clonotypes in MDS patients who responded to treatment, while non-responders had a higher frequency of contracted clonotypes following treatment. By applying GLIPH for antigen prediction, we found rare TCR specificity clusters shared by TCR clonotypes from different patients at pre- or following treatment. Our data show clear differences in TCR repertoires of MDS compared with healthy patients and that novel TCR clonotype emergence in response to HMA therapy was correlated with response. This suggests that response to HMA therapy may be partially driven by T-cell mediated immunity and that the immune-based therapies, which target the adaptive immune system, may play a significant role in select patients with MDS.","prettyUrl":"abbas-2021-fi","following":false,"created":"03/15/2021","featured":false,"publishedDate":"03/15/2021","urlOrId":"abbas-2021-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"5d8ddcab-5cd5-4a16-ac94-eb4a277c6831","title":"Bruton’s Tyrosine Kinase Supports Gut Mucosal Immunity and Commensal Microbiome Recognition in Autoimmune Arthritis","investigator":"Bonami, Rachel H.","investigatorInstitution":"Departments of Medicine, Pathology, Microbiology and Immunology, Vanderbilt University","publicationName":"Frontiers in Immunology","researchArea":"Autoimmune Disorders","prettyUrls":{"191":"bonami-2021-biorxiv"},"prettyUrlList":["bonami-2021-biorxiv"],"summary":"Bruton’s tyrosine kinase (Btk) deficiency preferentially eliminates autoreactive B cells while sparing normal humoral responses, but has not been studied in mucosal immunity. Commensal microbes are essential for arthritis in K/BxN mice, used here to examine how BTK-mediated signaling interfaces with the microbiome. Btk-deficient K/BxN mice were found to have small Peyer’s Patches with reduced germinal center and IgA+ B cells. Although lamina propria IgA+ plasma cells were numerically normal, intestinal IgA was low and IgA coating of commensal bacteria was reduced. IgA-seq showed a shift in microbes that are normally IgA-coated into the uncoated fraction in Btk-deficient mice. In this altered microbial milieau, the proportion of Parabacteroides distasonis was reduced in Btk-deficient K/BxN mice. To determine whether P. distasonis contributes to arthritis, it was reintroduced into antibiotic-protected K/BxN mice, where it restored disease. This suggests that P. distasonis’ inability to thrive in Btk-deficient mice may be a factor in disease protection. Thus, BTK supports normal intestinal IgA development, with downstream effects on the microbiome that may contribute to autoimmunity.","prettyUrl":"bonami-2021-biorxiv","following":false,"created":"03/15/2021","featured":false,"publishedDate":"03/15/2021","urlOrId":"bonami-2021-biorxiv","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"96f46797-219a-44bc-a98a-27e4ad7ea311","title":"Immune evolution from preneoplasia to invasive lung adenocarcinomas and underlying molecular features","investigator":"Zhang, Jianjun","investigatorInstitution":"Departments of Genomic Medicine and Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center","publicationName":"Nature Communications","researchArea":"Cancer","prettyUrls":{"190":"dejima-2021-nc"},"prettyUrlList":["dejima-2021-nc"],"summary":"Coming soon...","prettyUrl":"dejima-2021-nc","following":false,"created":"03/04/2021","featured":false,"publishedDate":"03/04/2021","urlOrId":"dejima-2021-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"4429931f-1d19-4a0b-a1b5-5c32c5d2a14d","title":"CD8+ T-cell repertoire in HLA class I-mismatched alloreactive immune response","investigator":"Bettens Florence","investigatorInstitution":"Hopitaux Universitaire de Genéve","publicationName":"Frontiers in Immunology","researchArea":"BMT / Stem Cell Transplant","prettyUrls":{"189":"studer-bettens-2020-fi"},"prettyUrlList":["studer-bettens-2020-fi"],"summary":"In transplantation, direct allorecognition is a complex interplay between T-cell receptors (TCR) and HLA molecules and their bound peptides expressed on antigen-presenting cells. In analogy to HLA mismatched hematopoietic stem cell transplantation (HSCT), the TCR CDR3β repertoires of alloreactive cytotoxic CD8+ responder T cells, defined by the cell surface expression of CD137 and triggered in vitro by HLA mismatched stimulating cells, were analyzed in different HLA class I mismatched combinations. The same HLA mismatched stimulatory cells induced very different repertoires in distinct but HLA identical responders. Likewise, stimulator cells derived from HLA identical donors activated CD8+ cells expressing very different repertoires in the same mismatched responder. To mimic in vivo inflammation, expression of HLA class l antigens was upregulated in vitro on stimulating cells by the inflammatory cytokines TNFα and IFNβ. The repertoires differed whether the same responder cells were stimulated with cells treated or not with both cytokines. In conclusion, the selection and expansion of alloreactive cytotoxic T-cell clonotypes expressing a very diverse repertoire is observed repeatedly despite controlling for HLA disparities and is significantly influenced by the inflammatory status. This makes prediction of alloreactive T-cell repertoires a major challenge in HLA mismatched HSCT.In conclusion, the selection and expansion of alloreactive cytotoxic T-cell clonotypes expressing a very diverse repertoire is observed repeatedly despite controlling for HLA disparities and is significantly influenced by the inflammatory status. This makes prediction of alloreactive T-cell repertoires a major challenge in HLA mismatched HSCT.","prettyUrl":"studer-bettens-2020-fi","following":false,"created":"10/29/2020","featured":false,"publishedDate":"03/02/2021","urlOrId":"studer-bettens-2020-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d0a85759-47a3-494f-b452-98c78844c3a3","title":"Lymphohematopoietic graft-versus-host responses promote mixed chimerism in patients receiving intestinal transplantation","investigator":"Fu, Jianing","investigatorInstitution":"Columbia Center for Translational Immunology, Department of Medicine","publicationName":"Journal of Clinical Investigation","researchArea":"Organ transplant","prettyUrls":{"188":"fu-2021-jci"},"prettyUrlList":["fu-2021-jci"],"summary":"In humans receiving intestinal transplantation, long-term multilineage blood chimerism often develops. Donor T cell macrochimerism (≥4%) frequently occurs without graft-versus-host disease (GVHD) and is associated with reduced rejection. Here we demonstrate that patients with macrochimerism had high graft-versus-host (GvH) to host-versus-graft (HvG) T cell clonal ratios in their allografts. These GvH clones entered the circulation, where their peak levels were associated with declines in HvG clones early post-transplant, suggesting that GvH reactions may contribute to chimerism and control HvG responses without causing GVHD. Consistently, donor-derived T cells, including GvH clones, and CD34+ hematopoietic stem and progenitor cells (HSPCs) were simultaneously detected in the recipients’ bone marrow (BM) >100 days post-transplant. Individual GvH clones appeared in ileal mucosa or blood before detection in recipient BM, consistent with an intestinal mucosal origin, where donor GvH-reactive T cells expanded early upon entry of recipient antigen-presenting cells into the graft. These results, combined cytotoxic single cell transcriptional profiles of donor T cells in recipient BM, suggest that tissue-resident GvH-reactive donor T cells migrated into the recipient circulation and BM, where they destroyed recipient hematopoietic cells and promoted engraftment of graft-derived HSPCs that maintain chimerism. These mechanisms suggest an approach to achieving intestinal allograft tolerance.","prettyUrl":"fu-2021-jci","following":false,"created":"02/17/2021","featured":false,"publishedDate":"03/02/2021","urlOrId":"fu-2021-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"bdb58f11-c513-4c72-8154-e071f69b33d7","title":"A subset of cytotoxic effector memory T cells enhances CAR T cell efficacy in a model of pancreatic ductal adenocarcinoma","investigator":"Konduri, Vanaja","investigatorInstitution":"Department of Pathology & Immunology, Baylor College of Medicine","publicationName":"Science Translational Medicine","researchArea":"Response to Therapeutic Agent","prettyUrls":{"187":"konduri-2021-stm"},"prettyUrlList":["konduri-2021-stm"],"summary":"In humans, the natural killer (NK) cell marker CD161 identifies several subsets of T cells, including a polyclonal CD8 αβ T cell receptor-expressing subset with characteristic specificity for tissue-localized viruses. This subset also displays enhanced cytotoxic and memory phenotypes. Here, we characterized this unique T cell subset and determined its potential suitability for use in chimeric antigen receptor (CAR) T cell therapy. In mice, gene expression profiling among the CD161-equivalent CD8+ T cell populations (CD8+NK1.1+) revealed substantial up-regulation of granzymes, perforin, killer lectin-like receptors, and innate signaling molecules in comparison to CD8+NK1.1- T cells. Adoptive transfer of CD8+NK1.1+ cells from previously exposed animals offered substantially enhanced protection and improved survival against melanoma tumors and influenza infection compared to CD8+NK1.1- cells. Freshly isolated human CD8+CD61+ T cells exhibited heightened allogeneic killing activity in comparison to CD8+CD61- T cells or total peripheral blood mononuclear cells (PBMCs). To determine whether this subset might improve the antitumor efficacy of CAR T cell therapy against solid tumors, we compared bulk PBMCs, CD8+CD161-, and CD8+CD161+ T cells transduced with a human epidermal growth factor receptor-2 (HER2)-specific CAR construct. In vitro, CD8+CD161+ CAR-transduced T cells killed HER2+ targets faster and with greater efficiency. Similarly, these cells mediated enhanced in vivo antitumor efficacy in xenograft models of HER2+ pancreatic ductal adenocarcinoma, exhibiting elevated expression of granzymes and reduced expression of exhaustion markers. These data suggest that this T cell subset presents an opportunity to improve CAR T cell therapy for the treatment of solid tumors.","prettyUrl":"konduri-2021-stm","following":false,"created":"02/23/2021","featured":false,"publishedDate":"02/23/2021","urlOrId":"konduri-2021-stm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"726e6200-7a43-4771-9204-bfd912720f64","title":"VJ segment usage of TCR-Beta repertoire in monozygotic cystic fibrosis twins","investigator":"Tümmler, Burkhard","investigatorInstitution":"Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany","publicationName":"Frontiers in Immunology","researchArea":"Basic Immunology","prettyUrls":{"186":"fischer-2021-fi"},"prettyUrlList":["fischer-2021-fi"],"summary":"Sixteen monozygotic cystic fibrosis (CF) twin pairs of whom 14 pairs were homozygous for the most common p.Phe508del CFTR mutation were selected from the European Cystic Fibrosis Twin and Sibling Study Cohort. The monozygotic twins were examined in their T cell receptor (TCR) repertoire in peripheral blood by amplicon sequencing of the CDR3 variable region of the ß-chain. The recruitment of TCR J and V genes for recombination and selection in the thymus showed a strong genetic influence in the CF twin cohort as indicated by the shortest Jensen-Shannon distance to the twin individual. Exceptions were the clinically most discordant and/or most severely affected twin pairs where clonal expansion probably caused by recurrent pulmonary infections overshadowed the impact of the identical genomic blueprint. In general the Simpson clonality was low indicating that the population of TCRß clonotypes of the CF twins was dominated by the naïve T-cell repertoire. Intrapair sharing of clonotypes was significantly more frequent among monozygotic CF twins than among pairs of unrelated CF patients. Complete nucleotide sequence identity was observed in about 0.11% of CDR3 sequences which partially should represent persisting fetal clones derived from the same progenitor T cells. Complete amino acid sequence identity was noted in 0.59% of clonotypes. Of the nearly 40,000 frequent amino acid clonotypes shared by at least two twin siblings 99.8% were already known within the immuneACCESS database and only 73 had yet not been detected indicating that the CDR3ß repertoire of CF children and adolescents does not carry a disease-specific signature but rather shares public clones with that of the non-CF community. Clonotypes shared within twin pairs and between unrelated CF siblings were highly abundant among healthy non-CF people, less represented in individuals with infectious disease and uncommon in patients with cancer. This subset of shared CF clonotypes defines CDR3 amino acid sequences that are more common in health than in disease.","prettyUrl":"fischer-2021-fi","following":false,"created":"02/09/2021","featured":false,"publishedDate":"02/15/2021","urlOrId":"fischer-2021-fi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"cbe97c48-8bc2-4aba-9028-50a3593eea44","title":"The immune suppressive microenvironment affects efficacy of radio-immunotherapy in brain metastasis","investigator":"Niesel, Katja","investigatorInstitution":"Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy","publicationName":"EMBO Molecular Medicine","researchArea":"Cancer","prettyUrls":{"185":"niesel-2021-embomm"},"prettyUrlList":["niesel-2021-embomm"],"summary":"The tumor microenvironment in brain metastases is characterized by high myeloid cell content associated with immune-suppressive and cancer-permissive functions. Moreover, brain metastases induce the recruitment of lymphocytes. Despite their presence, T cell-directed therapies fail to elicit effective anti-tumor immune responses. Here we seek to evaluate the applicability of radio-immunotherapy to modulate tumor immunity and overcome inhibitory effects that diminish anti-cancer activity. Radiotherapy-induced immune modulation resulted in an increase in cytotoxic T cell numbers and prevented the induction of lymphocyte-mediated immune suppression. Radio-immunotherapy led to significantly improved tumor control with prolonged median survival in experimental breast-to-brain metastasis. However, long-term efficacy was not observed. Recurrent brain metastases showed accumulation of blood-borne PD-L1+ myeloid cells after radio-immunotherapy indicating the establishment of an immune-suppressive environment to counteract re-activated T cell responses. This finding was further supported by transcriptional analyses indicating a crucial role for monocyte-derived macrophages in mediating immune-suppression and regulating T cell function. Therefore, selective targeting of immune suppressive functions of myeloid cells is expected to be critical for improved therapeutic efficacy of radio-immunotherapy in brain metastases.","prettyUrl":"niesel-2021-embomm","following":false,"created":"02/12/2021","featured":false,"publishedDate":"02/15/2021","urlOrId":"niesel-2021-embomm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b9c411fe-a1d7-4f2d-9df2-7450cf38cc42","title":"T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors","investigator":"Ito, Fumito","investigatorInstitution":"Roswell Park Comprehensive Cancer Center","publicationName":"Nature Communications","researchArea":"Response to Therapeutic Agent","prettyUrls":{"184":"yamauchi-icb-2021"},"prettyUrlList":["yamauchi-icb-2021"],"summary":"Immune checkpoint inhibitors (ICI) have revolutionized treatment for various cancers;\nhowever, durable response is limited to only a subset of patients. Discovery of blood-based biomarkers that reflect dynamic change of the tumor microenvironment, and predict response to ICI, will markedly improve current treatment regimens. Here, we investigate CX3C chemokine receptor 1 (CX3CR1), a marker of T-cell differentiation, as a predictive correlate of response to ICI therapy. Successful treatment of tumor-bearing mice with ICI increases the frequency and T-cell receptor clonality of the peripheral CX3CR1+CD8+ T-cell subset that includes an enriched repertoire of tumor-specific and tumor-infiltrating CD8+ T cells. Furthermore, an increase in the frequency of the CX3CR1+ subset in circulating CD8+ T cells early after initiation of anti-PD-1 therapy correlates with response and survival in patients with non-small cell lung cancer. Collectively, these data support T-cell CX3CR1 expression as a blood-based dynamic early on-treatment predictor of response to ICI therapy.","prettyUrl":"yamauchi-icb-2021","following":false,"created":"02/04/2021","featured":false,"publishedDate":"02/04/2021","urlOrId":"yamauchi-icb-2021","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"52dc9f7c-be6d-4421-aef7-e4d42915506d","title":"Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells","investigator":"Weinberg, Andrew","investigatorInstitution":"Earle A. Chiles Research Institute, Providence Cancer Institute","publicationName":"Nature Communications","researchArea":"Cancer Immunotherapy","prettyUrls":{"183":"duhen-2020-nc"},"prettyUrlList":["duhen-2020-nc"],"summary":"Despite the success of checkpoint blockade in some cancer patients, there is an unmet need to improve outcomes. Targeting alternative pathways, such as costimulatory molecules (e.g. OX40, GITR, and 4-1BB), can enhance T cell immunity in tumor-bearing hosts. Here we describe the results from a phase Ib clinical trial (NCT02274155) in which 17 patients with locally advanced head and neck squamous cell carcinoma (HNSCC) received a murine anti-human OX40 agonist antibody (MEDI6469) prior to definitive surgical resection. The primary endpoint was to determine safety and feasibility of the anti-OX40 neoadjuvant treatment. The secondary objective was to assess the effect of anti-OX40 on lymphocyte subsets in the tumor and blood. Neoadjuvant anti-OX40 was well tolerated and did not delay surgery, thus meeting the primary endpoint. Peripheral blood phenotyping data show increases in CD4+ and CD8+ T cell proliferation two weeks after anti-OX40 administration. Comparison of tumor biopsies before and after treatment reveals an increase of activated, conventional CD4+ tumor-infiltrating lymphocytes (TIL) in most patients and higher clonality by TCRb sequencing. Analyses of CD8+ TIL show increases in tumor-antigen reactive, proliferating CD103+CD39+ cells in 25% of patients with evaluable tumor tissue (N=4/16), all of whom remain disease-free.\nThese data provide evidence that anti-OX40 prior to surgery is safe and can increase activation and proliferation of CD4+ and CD8+ T cells in blood and tumor. Our work suggests that increases in the tumor-reactive CD103+CD39+ CD8+ TIL could serve as a potential biomarker of anti-OX40 clinical activity. These data provide evidence that anti-OX40 administered in the neoadjuvant setting is safe and can increase activation and proliferation of CD4+ and CD8+ T cells in blood and tumor. Our work suggests that increases in the tumor-reactive CD103+CD39+ CD8+ TIL could serve as a potential biomarker of anti-OX40 clinical activity.","prettyUrl":"duhen-2020-nc","following":false,"created":"02/02/2021","featured":false,"publishedDate":"02/02/2021","urlOrId":"duhen-2020-nc","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"063efde8-3071-43b9-b327-83e42ffa5ef2","title":"A vaccine targeting mutant IDH1 in newly diagnosed glioma","investigator":"Michael, Platten","investigatorInstitution":"CCU Neuroimmunology, German Cancer Research Center","publicationName":"Nature","researchArea":"Vaccine efficacy","prettyUrls":{"182":"platten-2021-nature"},"prettyUrlList":["platten-2021-nature"],"summary":"Mutated isocitrate dehydrogenase 1 (IDH1) defines a molecularly distinct subtype of diffuse glioma1-3. The most common IDH1 mutation in gliomas affects codon 132 and encodes IDH1(R132H), which harbours a shared clonal neoepitope that is presented on major histocompatibility complex (MHC) class II4,5. An IDH1(R132H)-specific peptide vaccine (IDH1-vac) induces specific therapeutic T helper cell responses that are effective against IDH1(R132H)+ tumours in syngeneic MHC-humanized mice4,6-8. Here we describe a multicentre, single-arm, open-label, first-in-humans phase I trial that we carried out in 33 patients with newly diagnosed World Health Organization grade 3 and 4 IDH1(R132H)+ astrocytomas (Neurooncology Working Group of the German Cancer Society trial 16 (NOA16), ClinicalTrials.gov identifier NCT02454634). The trial met its primary safety endpoint, with vaccine-related adverse events restricted to grade 1. Vaccine-induced immune responses were observed in 93.3% of patients across multiple MHC alleles. Three-year progression-free and death-free rates were 0.63 and 0.84, respectively. Patients with immune responses showed a two-year progression-free rate of 0.82. Two patients without an immune response showed tumour progression within two years of first diagnosis. A mutation-specificity score that incorporates the duration and level of vaccine-induced IDH1(R132H)-specific T cell responses was associated with intratumoral presentation of the IDH1(R132H) neoantigen in pre-treatment tumour tissue. There was a high frequency of pseudoprogression, which indicates intratumoral inflammatory reactions. Pseudoprogression was associated with increased vaccine-induced peripheral T cell responses. Combined single-cell RNA and T cell receptor sequencing showed that tumour-infiltrating CD40LG+ and CXCL13+ T helper cell clusters in a patient with pseudoprogression were dominated by a single IDH1(R132H)-reactive T cell receptor.","prettyUrl":"platten-2021-nature","following":false,"created":"02/02/2021","featured":false,"publishedDate":"02/02/2021","urlOrId":"platten-2021-nature","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"83168555-cff7-4274-bbf4-3fda70c75b4a","title":"Characterization of ascites- and tumor-infiltrating γδ T cells reveals distinct repertoires and a beneficial role in ovarian cancer","investigator":"Foord, Emelie","investigatorInstitution":"Department of Clinical Science, Intervention and Technology","publicationName":"Science Translational Medicine","researchArea":"Cancer","prettyUrls":{"181":"foord-2020-stm"},"prettyUrlList":["foord-2020-stm"],"summary":"The role of γδ T cells in antitumor immunity has been under investigation for the past two decades, but little is known about their contribution to clinical outcomes in patients. Here, we set out to define the clonotypic, phenotypic, and functional features of γδ T cells in peripheral blood, ascites, and metastatic tumor tissue from patients with advanced epithelial ovarian cancer. T cell receptor (TCR) sequencing of the γ chain revealed that tumor-infiltrating γδ T cells have a unique and skewed repertoire with high TCR diversity and low clonality. In contrast, ascites-derived γδ T cells presented a lower TCR diversity and higher clonality, suggesting a TCR-dependent clonal focusing at this site. Further investigation showed that tumor samples had abundant γδ T cells with a tissue-resident, activation-associated phenotype, less usage of Vγ9 and an impaired response to adaptive-associated stimuli, implying an innate-like activation pathway, rather than an adaptive TCR-engaging pathway, at these tumor sites. Furthermore, high γδ T cell cytokine responsiveness upon stimulation was associated with a favorable outcome for patients in terms of both overall survival and reduced residual tumor burden after primary surgery. Last, the functionality of γδ T cells and patient survival were negatively affected by the proportions of CD39-expressing T cells, highlighting the potential of CD39 as a target to improve γδ T cell responses and unleash their antitumor capabilities.","prettyUrl":"foord-2020-stm","following":false,"created":"11/02/2020","featured":false,"publishedDate":"01/21/2021","urlOrId":"foord-2020-stm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"c590809f-e88e-4310-842d-82659bc480c5","title":"Safety and efficacy of consolidation therapy with ipilimumab plus nivolumab after autologous stem cell transplantation","investigator":"Skarbnik, Alan P","investigatorInstitution":"Novant Health Cancer Institute","publicationName":"Transplantation and Cellular Therapy","researchArea":"Cancer Immunotherapy","prettyUrls":{"180":"skarbnik-2020-tct"},"prettyUrlList":["skarbnik-2020-tct"],"summary":"Background: Autologous hematopoietic stem cell transplantation (ASCT) is a standard-of-care treatment for many hematological malignancies. Progression of disease after ASCT is the primary cause of treatment failure. \n\nObjective: In this Phase Ib trial, we studied the safety and clinical effect of combined checkpoint inhibition with ipilimumab and nivolumab as a consolidation strategy after ASCT for patients with high-risk diffuse large B-cell Lymphoma (DLBCL), mature T-cell lymphoma (TCL) and multiple myeloma (MM). Study Design: Starting 14-28 days after ASCT, patients received ipilimumab (1 mg/kg, intravenous, on day 1 of weeks 1, 4, 7, 10, 16, 22) and nivolumab (3 mg/kg, intravenous, on day 1 of weeks 1, 4, 7, 10, 12, 14, 16, 18, 20, 22, 24, 26). \n\nResults: Patients received a median of five doses of ipilimumab and eight doses of nivolumab. Thirty-five patients were included in the intent-to-treat population. Ninety-four percent of patients experienced immune-related adverse events (irAEs) of any grade. Ninety-seven percent of irAEs resolved spontaneously or by holding study drugs and instituting high-dose corticosteroid therapy. Progression-free and overall survival at 18 months post-ASCT for each disease cohort were, respectively: primary refractory DLBCL, 85.7% and 100%; relapsed DLBCL, 28.6% and 57.1%; frontline TCL: Not Evaluable and 80%; relapsed TCL, 25% and 75%; high-risk transplantnaïve MM, 57.1% and 87% and MM relapsed within 3 years of first ASCT, 40% and 100%. \n\nConclusion: We conclude that combined checkpoint inhibition appears tolerable as a consolidation strategy after ASCT and in addition to the potential clinical efficacy observed in some subsets of disease, T cell receptor repertoire, Tregulatory phenotype and gut microbiota profiles provide a biologic rationale warranting further study of this approach.","prettyUrl":"skarbnik-2020-tct","following":false,"created":"01/08/2021","featured":false,"publishedDate":"01/15/2021","urlOrId":"skarbnik-2020-tct","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"a97ed8a3-ca37-46c4-b449-bf119dfc9d8a","title":"Distinct T cell receptor repertoire diversity of clinically defined high-grade serous ovarian cancer treatment subgroups","investigator":"Lee, Sanghoon","investigatorInstitution":"Department of Systems Biology, The University of Texas MD Anderson Cancer Center","publicationName":"iScience","researchArea":"Cancer","prettyUrls":{"179":"sanghoon-2021-iscience"},"prettyUrlList":["sanghoon-2021-iscience"],"summary":"In patients with high-grade serous ovarian cancer (HGSC), it is unclear which genomic features are related to complete gross resection (R0), which is typically associated with better clinical outcomes, or response to neoadjuvant chemotherapy (NACT). In this study, we evaluated T cell receptor (TCR) repertoire diversity in primary and metastatic tumor samples (n=90) from clinically well-annotated patients with HGSC who achieved R0 or received NACT with excellent or poor response based on a laparoscopic triage algorithm. TCR sequencing followed by an integrative analysis with comprehensive multi-omics data identified higher TCR diversity (e.g., higher number of unique productive sequences and less clonal relatedness) in the R0 group than in the NACT group. We found enrichment of specific TCRβ genes usage, as well as distinct mutual exclusiveness and co-occurrence pattern of TCRβ genes among the groups. We also found significantly positive correlations between clonal relatedness and neoantigens and between copy number variations and mutation load in the groups.","prettyUrl":"sanghoon-2021-iscience","following":false,"created":"06/09/2020","featured":false,"publishedDate":"01/11/2021","urlOrId":"sanghoon-2021-iscience","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"08ff60f1-9b49-4637-a328-1a6590f0c4f3","title":"Resident and circulating memory T cells persist for years in melanoma patients with durable responses to immunotherapy","investigator":"Turk, Mary Jo","investigatorInstitution":"Geisel School of Medicine at Dartmouth","publicationName":"Nature Cancer","researchArea":"Cancer Immunotherapy","prettyUrls":{"178":"han-2021-natcancer"},"prettyUrlList":["han-2021-natcancer"],"summary":"While T-cell responses to cancer immunotherapy have been avidly studied, long-lived 34 memory has been poorly characterized. In a cohort of metastatic melanoma survivors with exceptional responses to immunotherapy, we probed memory CD8+ 35 T-cell responses across 36 tissues, and across several years. Single-cell RNA sequencing revealed three subsets of resident 37 memory T (TRM) cells shared between tumors and distant vitiligo-affected skin. Paired T-cell 38 receptor sequencing further identified clonotypes in tumors that co-existed as TRM in skin and as 39 effector memory T (TEM) cells in blood. Clonotypes that dispersed throughout tumor, skin, and 40 blood preferentially expressed a IFNG / TNF-high signature, which had a strong prognostic value 41 for melanoma patients. Remarkably, clonotypes from tumors were found in patient skin and 42 blood up to nine years later, with skin maintaining the most focused tumor-associated clonal 43 repertoire. These studies reveal that cancer survivors can maintain durable memory as functional, 44 broadly-distributed TRM and TEM compartments.","prettyUrl":"han-2021-natcancer","following":false,"created":"01/05/2021","featured":false,"publishedDate":"01/06/2021","urlOrId":"han-2021-natcancer","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"3c521221-d6b2-4ea3-b8f6-cc7869e5e6f5","title":"Broadly reactive human CD4+ T cells against Enterobacteriaceae are found in the naïve repertoire and are clonally expanded in the memory repertoire","investigator":"Sallusto, Federica","investigatorInstitution":"Institute for Research in Biomedicine","publicationName":"European Journal of Immunology","researchArea":"Basic Immunology","prettyUrls":{"177":"cassotta-2020-eji"},"prettyUrlList":["cassotta-2020-eji"],"summary":"Enterobacteriaceae are a large family of Gram-negative bacteria that includes both commensals and opportunistic pathogens. The latter can cause severe nosocomial infections, with outbreaks of multi-antibiotics resistant strains, thus being a major public health threat. In this study, we report that Enterobacteriaceae-reactive memory Th cells were highly enriched in a CCR6+ CXCR3+ Th1*/17 cell subset and produced IFN-γ, IL-17A, and IL-22. This T cell subset was severely reduced in septic patients with K. pneumoniae bloodstream infection who also selectively lacked circulating K. pneumonie-reactive T cells. By combining heterologous antigenic stimulation, single cell cloning and TCR Vβ sequencing, we demonstrate that a large fraction of memory Th cell clones was broadly cross-reactive to several Enterobacteriaceae species. These cross-reactive Th cell clones were expanded in vivo and a large fraction of them recognized the conserved outer membrane protein A antigen. Interestingly, Enterobacteriaceae broadly cross-reactive T cells were also prominent among in vitro primed naïve T cells. Collectively, these data point to the existence of immunodominant T cell epitopes shared among different Enterobacteriaceae species and targeted by cross-reactive T cells that are readily found in the pre-immune repertoire and are clonally expanded in the memory repertoire.","prettyUrl":"cassotta-2020-eji","following":false,"created":"01/04/2021","featured":false,"publishedDate":"01/04/2021","urlOrId":"cassotta-2020-eji","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"41bd4c0f-4675-40f4-9a31-8bfcaae8a785","title":"Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial","investigator":"Cascone, Tina","investigatorInstitution":"Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center","publicationName":"Nature Medicine","researchArea":"Other","prettyUrls":{"176":"cascone-2020-nm"},"prettyUrlList":["cascone-2020-nm"],"summary":"Ipilimumab improves clinical outcomes when combined with nivolumab in metastatic non-small cell lung cancer (NSCLC), but its efficacy and impact on the immune microenvironment in operable NSCLC remain unclear. We report the results of the phase 2 randomized NEOSTAR trial (NCT03158129) of neoadjuvant nivolumab or nivolumab + ipilimumab followed by surgery in 44 patients with operable NSCLC, using major pathologic response (MPR) as the primary endpoint. The MPR rate for each treatment arm was tested against historical controls of neoadjuvant chemotherapy. The nivolumab + ipilimumab arm met the prespecified primary endpoint threshold of 6 MPRs in 21 patients, achieving a 38% MPR rate (8/21). We observed a 22% MPR rate (5/23) in the nivolumab arm. In 37 patients resected on trial, nivolumab and nivolumab + ipilimumab produced MPR rates of 24% (5/21) and 50% (8/16), respectively. Compared with nivolumab, nivolumab + ipilimumab resulted in higher pathologic complete response rates (10% versus 38%), less viable tumor (median 50% versus 9%), and greater frequencies of effector, tissue-resident memory and effector memory T cells. Increased abundance of gut Ruminococcus and Akkermansia spp. was associated with MPR to dual therapy. Our data indicate that neoadjuvant nivolumab + ipilimumab-based therapy enhances pathologic responses, tumor immune infiltrates and immunologic memory, and merits further investigation in operable NSCLC.","prettyUrl":"cascone-2020-nm","following":false,"created":"09/21/2020","featured":false,"publishedDate":"12/10/2020","urlOrId":"cascone-2020-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7b134d46-565b-4c06-bb4b-d72faaa64881","title":"Antigen-driven clonal selection shapes the persistence of HIV-1 infected CD4+ T cells in vivo","investigator":"Simonetti, Francesco R.","investigatorInstitution":"Department of Medicine, Johns Hopkins University School of Medicine","publicationName":"Journal of Clinical Investigation","researchArea":"HIV","prettyUrls":{"175":"simonetti-2020-jci"},"prettyUrlList":["simonetti-2020-jci"],"summary":"Clonal expansion of infected CD4+ T cells is a major mechanism of HIV-1 persistence and a barrier to cure. Potential causes are homeostatic proliferation, effects of HIV-1 integration, and interaction with antigens. Here we show that it is possible to link antigen responsiveness, full proviral sequence, integration site, and T cell receptor β-chain (TCRβ) sequence to examine the role of recurrent antigenic exposure in maintaining the HIV-1 reservoir. We isolated Cytomegalovirus (CMV)- and Gag-responding CD4+ T cells from 10 treated individuals. Proviral populations in CMV-responding cells were dominated by large clones, including clones harboring replication-competent proviruses. TCRβ repertoires showed high clonality driven by converging adaptive responses. Although some proviruses were in genes linked to HIV-1 persistence (BACH2, STAT5B, MKL1), proliferation of infected cells under antigenic stimulation occurred regardless of the site of integration. Paired TCRβ-integration site analysis showed that infection could occur early or late in the course of a clone’s response to antigen and could generate infected cell populations too large to be explained solely by homeostatic proliferation. Together these findings implicate antigen-driven clonal selection as a major factor in HIV-1 persistence, a finding that will be a difficult challenge to eradication efforts.","prettyUrl":"simonetti-2020-jci","following":false,"created":"11/20/2020","featured":false,"publishedDate":"12/07/2020","urlOrId":"simonetti-2020-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"407e3aa0-76ad-4286-a8da-555c9a845b1c","title":"Adult-Onset Anti-Citrullinated Peptide Antibody-Negative Destructive Rheumatoid Arthritis Is Characterized by a Disease-Specific CD8+ T Lymphocyte Signature","investigator":"Mustjoki, Satu","investigatorInstitution":"University of Helsinki","publicationName":"Frontiers in Immunology","researchArea":"Autoimmune Disorders","prettyUrls":{"174":"kelkka-2020-jci"},"prettyUrlList":["kelkka-2020-jci"],"summary":"Rheumatoid arthritis (RA) is a complex autoimmune disease targeting synovial joints. Traditionally, RA is divided into seropositive (SP) and seronegative (SN) disease forms, the latter consisting of an array of unrelated diseases with joint involvement. Recently, we described a severe form of SN-RA that associates with characteristic joint destruction. Here, we sought biological characteristics to differentiate this rare but aggressive anti-citrullinated peptide antibody-negative destructive RA (CND-RA) from early seropositive (SP-RA) and seronegative rheumatoid arthritis (SN-RA). We also aimed to study cytotoxic CD8+ lymphocytes in autoimmune arthritis.\n \nCND-RA, SP-RA and SN-RA were compared to healthy controls to reveal differences in T-cell receptor beta (TCRβ) repertoire, cytokine levels and autoantibody repertoires. Whole-exome sequencing (WES) followed by single-cell RNA-sequencing (sc-RNA-seq) was performed to study somatic mutations in a clonally expanded CD8+ lymphocyte population in an index patient. A unique TCRβ signature was detected in CND-RA patients. In addition, CND-RA patients expressed higher levels of the bone destruction-associated TNFSF14 cytokine. Blood IgG repertoire from CND-RA patients recognized fewer endogenous proteins than SP-RA patients’ repertoires. Using WES, we detected a stable mutation profile in the clonally expanded CD8+ T-cell population characterized by cytotoxic gene expression signature discovered by sc-RNA-sequencing.\n \nOur results identify CND-RA as an independent RA subset and reveal a CND-RA specific TCR signature in the CD8+ lymphocytes. Improved classification of seronegative RA patients underlines the heterogeneity of RA and also, facilitates development of improved therapeutic options for the treatment resistant patients.","prettyUrl":"kelkka-2020-jci","following":false,"created":"11/13/2020","featured":false,"publishedDate":"11/23/2020","urlOrId":"kelkka-2020-jci","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"946757e6-58e2-4235-b011-58f2599f04e2","title":"Human thymic T cell repertoire is imprinted with strong convergence to shared sequences","investigator":"Arstila, T Petteri","investigatorInstitution":"University of Helsinki","publicationName":"Molecular Immunology","researchArea":"Basic Immunology","prettyUrls":{"173":"heikkila-2020-mi"},"prettyUrlList":["heikkila-2020-mi"],"summary":"A highly diverse repertoire of T cell antigen receptors (TCR) is created in the thymus by recombination of gene segments and the insertion or deletion of nucleotides at the junctions. Using next-generation TCR sequencing we define here the features of recombination and selection in the human TCRα and TCRβ locus, and show that a strikingly high proportion of the repertoire is shared by unrelated individuals. The thymic TCRα nucleotide repertoire was more diverse than TCRβ, with 4.1 × 106 vs. 0.81 × 106 unique clonotypes, and contained nonproductive clonotypes at a higher frequency (69.2% vs. 21.2%). The convergence of distinct nucleotide clonotypes to the same amino acid sequences was higher in TCRα than in TCRβ repertoire (1.45 vs. 1.06 nucleotide sequences per amino acid sequence in thymus). The gene segment usage was biased, and generally all individuals favored the same genes in both TCRα and TCRβ loci. Despite the high diversity, a large fraction of the repertoire was found in more than one donor. The shared fraction was bigger in TCRα than TCRβ repertoire, and more common in in-frame sequences than in nonproductive sequences. Thus, both biases in rearrangement and thymic selection are likely to contribute to the generation of shared repertoire in humans.","prettyUrl":"heikkila-2020-mi","following":false,"created":"11/16/2020","featured":false,"publishedDate":"11/16/2020","urlOrId":"heikkila-2020-mi","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"60e6773f-9b9f-47e6-b33a-1532cdc630e3","title":"Exhaustion of tumour-infiltrating T-cell receptor repertoire diversity is an age-dependent indicator of immunological fitness independently predictive of clinical outcome in Burkitt lymphoma","investigator":"Gebauer, Niklas","investigatorInstitution":"Klinik für Hämatologie und Onkologie UKSH Campus Lübeck","publicationName":"British Journal of Haematology","researchArea":"Cancer","prettyUrls":{"172":"rieken-2020-bjh"},"prettyUrlList":["rieken-2020-bjh"],"summary":"Burkitt-lymphoma (BL) is an aggressive-B-cell-malignancy derived from germinal-centre B-cells. Curative therapy traditionally requires intensive immunochemotherapy. Recently, immuno-oncological approaches, modulating the T-cell tumour-response, were approved for the treatment of a variety of malignancies. The architecture of the tumour-infiltrating T-cell-receptor (TCR)-repertoire in BL remains insufficiently characterized.\nWe therefore performed a large-scale, next-generation-sequencing study of the CDR3-region of the TCRβ chain-repertoire in a large cohort of all epidemiological subtypes of BL (n=82) and diffuse large B-cell-lymphoma (DLBCL; n=34). Molecular data were subsequently assessed for correlation with clinical outcome. \nOur investigations revealed an age-dependent immunoprofile in BL similar to DLBCL. Moreover, we found several public clonotypes in numerous patients suggestive of shared tumour-neoantigen selection exclusive to BL and distinct from DLBCL regardless of EBV and/or HIV-status. Compared with baseline, longitudinal analysis unveiled significant repertoire restrictions upon relapse (p=0.0437) while productive-TCR-repertoire clonality proved as a useful indicator of both overall and progression-free-survival (OS: p=0.0001; HR: 6.220; CI: 2.263–11.78; PFS: p=0.0025; HR: 3.086; CI: 1.555–7.030). Multivariate-analysis confirmed its independence from established prognosticators, including age at diagnosis and comorbidities.\nOur findings establish the clinical relevance of the architecture and clonality of the TCR-repertoire and its age-determined dynamics in BL.","prettyUrl":"rieken-2020-bjh","following":false,"created":"11/12/2020","featured":false,"publishedDate":"11/13/2020","urlOrId":"rieken-2020-bjh","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"7f60f599-1c4b-4ac7-aca0-d8f30759d05f","title":"Expansion of Human Papillomavirus (HPV)-Specific T Cells in Periphery and Cervix in a Therapeutic Vaccine Recipient Whose Cervical High-Grade Squamous Intraepithelial Lesion Regressed","investigator":"Nakagawa, Mayumi","investigatorInstitution":"Department of Pathology, University of Arkansas for Medical Sciences","publicationName":"Frontiers in Immunology","researchArea":"Response to Therapeutic Agent","prettyUrls":{"171":"shibata-2020-hpv"},"prettyUrlList":["shibata-2020-hpv"],"summary":"Advances in high-throughput sequencing have revolutionized the manner with which we can study T cell responses. We describe a woman who received a human papillomavirus (HPV) therapeutic vaccine called PepCan, and experienced complete resolution of her cervical high-grade squamous intraepithelial lesion. By performing bulk T cell receptor (TCR) β deep sequencing of peripheral blood mononuclear cells before and after 4 vaccinations, 70 putatively vaccine-specific clonotypes were identified for being significantly increased using a beta-binomial model. In order to verify the vaccine-specificity of these clonotypes, T cells with specificity to a region, HPV 16 E6 91-115, previously identified to be vaccine-induced using an interferon-γ enzyme-linked immunospot assay, were sorted and analyzed using single-cell RNA-seq and TCR sequencing. HPV specificity in 60 of the 70 clonotypes identified to be vaccine-specific was demonstrated. TCR β bulk sequencing of the cervical liquid-based cytology samples and cervical formalin-fixed paraffin-embedded samples before and after 4 vaccinations demonstrated the presence of these HPV-specific T cells in the cervix. Combining traditional and cutting-edge immunomonitoring techniques enabled us to demonstrate expansion of HPV-antigen specific T cells not only in the periphery but also in the cervix. Such an approach should be useful as a novel approach to assess vaccine-specific responses in various anatomical areas.","prettyUrl":"shibata-2020-hpv","following":false,"created":"10/30/2020","featured":false,"publishedDate":"11/10/2020","urlOrId":"shibata-2020-hpv","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"eaddbb33-f612-49a5-975f-e5d1daba35af","title":"Permissive HLA-DPB1 mismatches in HCT depend on immunopeptidome divergence and editing by HLA-DM","investigator":"Meurer, Thuja","investigatorInstitution":"Institute for Experimental Cellular Therapy","publicationName":"Blood","researchArea":"BMT / Stem Cell Transplant","prettyUrls":{"170":"meurer-2020-blood"},"prettyUrlList":["meurer-2020-blood"],"summary":"In hematopoietic cell transplantation (HCT), permissive HLA-DPB1 mismatches between patients and their unrelated donors (UD) are associated with improved outcomes compared to non-permissive mismatches, but the underlying mechanism is incompletely understood. Here we used mass spectrometry, T-cell receptor-beta (TCR) deep sequencing, and cellular in vitro models of alloreactivity to interrogate the HLA-DP immunopeptidome and its role in alloreactive T cell responses. We find that permissive HLA-DPB1 mismatches display significantly higher peptide repertoire overlaps compared to their non-permissive counterparts, resulting in lower frequency and diversity of alloreactive TCR clonotypes in healthy individuals and transplanted patients. Permissiveness can be reversed by the absence of the peptide editor HLA-DM, or the presence of its antagonist HLA-DO, through significant broadening of the peptide repertoire. Our data establish the degree of immunopeptidome divergence between donor and recipient as the mechanistic basis for the clinically relevant permissive HLA-DPB1 mismatches in HCT, and show that permissiveness is dependent on HLA-DM-mediated peptide editing. Its key role for harnessing T-cell alloreactivity to HLA-DP highlights HLA-DM as a potential novel target for cellular and immunotherapy of leukemia.","prettyUrl":"meurer-2020-blood","following":false,"created":"11/09/2020","featured":false,"publishedDate":"11/09/2020","urlOrId":"meurer-2020-blood","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"08e519bc-34a9-4462-8abf-a1c5b957a472","title":"Long-term Sculpting of the B-cell Repertoire Following Cancer Immunotherapy in Patients Treated with Sipuleucel-T","investigator":"Zhang, Li","investigatorInstitution":"Departments of Medicine and Epidemiology & Biostatistics, University of California San Francisco","publicationName":"Cancer Immunology Research","researchArea":"Cancer Immunotherapy","prettyUrls":{"168":"zhang-2020-cir"},"prettyUrlList":["zhang-2020-cir"],"summary":"Sipuleucel-T is an autologous cellular immunotherapy, administered as up to 3 infusions, for metastatic castration-resistant prostate cancer (mCRPC). Sipuleucel-T induces T- and B-cell responses to prostatic acid phosphatase (PAP) that correlate with improved survival. The long-term impact, however, of sipuleucel-T on tumor antigen-specific immunologic memory remains unknown. In particular, B cell responses as measured by antigen-specific antibody responses and B cell receptor (BCR) sequences remain unknown. To evaluate whether sipuleucel-T can induce long-term immunologic memory, we examined circulating B cell responses before and after sipuleucel-T treatment in two groups of mCRPC patients: those who had previously received sipuleucel-T (treated; median, 8.9 years since the previous treatment) versus those who had not (naïve). Before re-treatment, previously treated patients exhibited persistent antibody responses as well as more focused and convergent BCR repertoires with distinct V(D)J gene usage compared with naïve patients. After retreatment, previously treated patients maintained high frequency clones and developed more convergent BCRs at earlier time points unlike naive patients. With the first sipuleucel-T infusion specifically, previously-treated patients had less shuffling within the 100-most abundant baseline clones whereas, in contrast, naïve patients exhibited great BCR turnover with a continued influx of new B cell clones. Social network analysis showed that previously treated patients had more highly organized B cell repertoires, consistent with greater clonal maturation. Further, higher treatment-induced BCR clonality was correlated with longer survival for naïve patients. These results demonstrate the capacity of sipuleucel-T to induce long-term immune memory and lasting changes to the B cell repertoire.","prettyUrl":"zhang-2020-cir","following":false,"created":"10/21/2020","featured":false,"publishedDate":"10/22/2020","urlOrId":"zhang-2020-cir","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"71a8dfd3-8611-4d67-9c79-deefc6db37be","title":"immunoSEQ hsTCRB-V4b Control Data","investigator":"Hamm, David","investigatorInstitution":"Adaptive Biotechnologies","publicationName":"","researchArea":"Controls","prettyUrls":{"169":"tcrbv4-control"},"prettyUrlList":["tcrbv4-control"],"summary":"This data set features 158 Samples with Tissue Source, Indication and Race/Ethnicity information as tags, run on the immunoSEQ hsTCRBv4b Assay. There are 147 peripheral samples (58 PBMC from cancer patients and 88 blood from healthy controls) and 11 FFPE samples (both lung cancer and melanoma).\n\nBlood and PBMC samples were run as deeps with up to 6 ug of DNA for blood samples and up to 2 ug of total DNA for PBMCs.","prettyUrl":"tcrbv4-control","following":false,"created":"10/21/2020","featured":false,"publishedDate":"10/21/2020","urlOrId":"tcrbv4-control","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"1a04eec5-9219-443d-8d72-d2f49ff5e102","title":"CRISPR-Cas9-Edited Tacrolimus-Resistant Antiviral T Cells for Advanced Adoptive Immunotherapy in Transplant Recipients","investigator":"Schmueck-Henneresse, Michael","investigatorInstitution":"Charité University Medicine","publicationName":"Molecular Therapy","researchArea":"Organ transplant","prettyUrls":{"167":"amini-2020-mt"},"prettyUrlList":["amini-2020-mt"],"summary":"Viral infections, such as with cytomegalovirus (CMV), remain a major risk factor for mortality and morbidity of transplant recipients because of their requirement for lifelong immunosuppression (IS). Antiviral drugs often cause toxicity and sometimes fail to control disease. Thus, regeneration of the antiviral immune response by adoptive antiviral T cell therapy is an attractive alternative. Our recent data, however, show only short-term efficacy in some solid organ recipients, possibly because of malfunction in transferred T cells caused by ongoing IS. We developed a vector-free clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-based good manufacturing practice (GMP)-compliant protocol that efficiently targets and knocks out the gene for the adaptor protein FK506-binding protein 12 (FKBP12), required for the immunosuppressive function of tacrolimus. This was achieved by transient delivery of ribonucleoprotein complexes into CMV-specific T cells by electroporation. We confirmed the tacrolimus resistance of our gene-edited T cell products in vitro and demonstrated performance comparable with non-tacrolimus-treated unmodified T cells. The alternative calcineurin inhibitor cyclosporine A can be administered as a safety switch to shut down tacrolimus-resistant T cell activity in case of adverse effects. Furthermore, we performed safety assessments as a prerequisite for translation to first-in-human applications.","prettyUrl":"amini-2020-mt","following":false,"created":"09/21/2020","featured":false,"publishedDate":"09/21/2020","urlOrId":"amini-2020-mt","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d8fb6de2-d364-4b06-a0d7-0363eb0587cb","title":"Long-term robustness of a T-cell system emerging from somatic rescue of a genetic block in T-cell development","investigator":"Ehl, Stephan","investigatorInstitution":"Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI) and Center for Pediatric and Adolescent Medicine, Medical Center","publicationName":"EBioMedicine","researchArea":"Basic Immunology","prettyUrls":{"166":"kury-2020-ebiomed"},"prettyUrlList":["kury-2020-ebiomed"],"summary":"Backgound: The potential of a single progenitor cell to establish and maintain long-term protective T-cell immunity in humans is unknown. For genetic disorders disabling T-cell immunity, somatic reversion was shown to support limited T-cell development attenuating the clinical phenotype. However, the cases reported so far deteriorated over time leaving unanswered the important question of long-term activity of revertant precursors and the robustness of the resulting T-cell system.\n\nMethods: We applied TCRβ-CDR3 sequencing and mass cytometry on serial samples of a now 18 year-old SCIDX1 patient with somatic reversion to analyse the longitudinal diversification and stability of a T-cell system emerging from somatic gene rescue. \n\nFindings: We detected close to 105 individual CDR3β sequences in the patient. Blood samples of equal size contained about 10-fold fewer unique CDR3β sequences compared to healthy donors, indicating a surprisingly broad repertoire. Despite dramatic expansions and contractions of individual clonotypes representing up to 30% of the repertoire, stable diversity indices revealed that these transient clonal distortions did not cause long-term repertoire imbalance. Phenotypically, the T-cell system did not show evidence for progressive exhaustion. Combined with immunoglobulin substitution, the limited T-cell system in this patient supported an unremarkable clinical course over 18 years.\n\nInterpretation: Genetic correction in the appropriate cell type, in our patient most likely in a T-cell biased self-renewing hematopoietic progenitor, can yield a diverse T-cell system that provides long-term repertoire stability, does not show evidence for progressive exhaustion and is capable of providing protective and regulated T-cell immunity for at least two decades.","prettyUrl":"kury-2020-ebiomed","following":false,"created":"09/09/2020","featured":false,"publishedDate":"09/09/2020","urlOrId":"kury-2020-ebiomed","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"e5a53393-8bd9-4978-ae4d-84931c7c4ca6","title":"Radiation induces dynamic changes to the T cell repertoire in renal cell carcinoma patients","investigator":"Chow, Jacky","investigatorInstitution":"Roswell Park Comprehensive Cancer Center","publicationName":"PNAS","researchArea":"Cancer","prettyUrls":{"165":"chow-2020-pnas"},"prettyUrlList":["chow-2020-pnas"],"summary":"Clinical studies combining radiation and immunotherapy have shown promising response rates, strengthening efforts to sensitize tumors to immune-mediated attack. Thus, there is an ongoing surge in trials using preconditioning regimens with immunotherapy. Yet, due to the scarcity of resected tumors treated in situ with radiotherapy, there has been little investigation of radiation’s sole contributions to local and systemic antitumor immunity in patients. Without this access, translational studies have been limited to evaluating circulating immune subsets and systemic remodeling of peripheral T cell receptor repertoires. This constraint has left gaps in how radiation impacts intratumoral responses and whether tumor-resident T cell clones are amplified following treatment. Therefore, to interrogate the immune impact of radiation on the tumor microenvironment and test the hypothesis that radiation initiates local and systemic expansion of tumor-resident clones, we analyzed renal cell carcinomas from patients treated with stereotactic body radiation therapy. Transcriptomic comparisons were evaluated by bulk RNA sequencing. T cell receptor sequencing monitored repertoires during treatment. Pathway analysis showed radiation-specific enrichment of immune-related processes, and T cell receptor sequencing revealed increased clonality in radiation-treated tumors. The frequency of identified, tumor-enriched clonotypes was tracked across serial blood samples. We observed increased abundance of tumor-enriched clonotypes at 2 wk postradiation compared with pretreatment levels; however, this expansion was not sustained, and levels contracted toward baseline by 4 wk post treatment. Taken together, these results indicate robust intratumoral immune remodeling and a window of tumor-resident T cell expansion following radiation that may be leveraged for the rational design of combinatorial strategies.","prettyUrl":"chow-2020-pnas","following":false,"created":"08/21/2020","featured":false,"publishedDate":"09/08/2020","urlOrId":"chow-2020-pnas","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"b26fb32b-6add-49bc-8523-9f49eb930762","title":"A Peripheral Immune Signature of Responsiveness to PD-1 Blockade in Patients with Classical Hodgkin Lymphoma","investigator":"Cader, Fathima Zumla","investigatorInstitution":"Department of Medical Oncology","publicationName":"Nature Medicine","researchArea":"Cancer","prettyUrls":{"163":"cader-2020-nm"},"prettyUrlList":["cader-2020-nm"],"summary":"PD-1 blockade is highly effective in classical Hodgkin lymphomas (cHLs) which exhibit frequent chromosome 9p24.1/CD274 (PD-L1)/PDC1LG2 (PD-L2) copy gains. However, in this largely MHC class I-negative tumor, the mechanism of action of anti-PD-1 therapy remains undefined. We utilized the complementary approaches of T-cell receptor (TCR) sequencing and cytometry by time-of-flight analysis (CyTOF) to obtain a peripheral immune signature of responsiveness to PD-1 blockade in 56 patients treated in the CheckMate 205 phase II clinical trial (NCT02181738). Anti PD-1 therapy was most effective in patients with a diverse baseline TCR repertoire and an associated expansion of singleton clones during treatment. CD4+, but not CD8+, TCR diversity significantly increased during therapy, most strikingly in patients who achieved complete responses. Additionally, responding patients had an increased abundance of activated NK cells and a newly identified CD3-CD68+CD4+GrB+ subset. These studies highlight the roles of recently expanded, clonally diverse CD4+ T cells and innate effectors in the efficacy of PD-1 blockade in cHL.","prettyUrl":"cader-2020-nm","following":false,"created":"08/04/2020","featured":false,"publishedDate":"08/05/2020","urlOrId":"cader-2020-nm","species":null,"loci":null,"tags":null,"tagCategories":[]},{"projectId":"d47e5ad9-51b8-4f78-9a30-1b615ff36bde","title":"A large-scale database of T-cell receptor beta (TCRb) sequences and binding associations from natural and synthetic exposure to SARS-CoV-2","investigator":"Robins, Harlan","investigatorInstitution":"Adaptive Biotechnologies","publicationName":"","researchArea":"Infectious Disease","prettyUrls":{"162":"covid-2020"},"prettyUrlList":["covid-2020"],"summary":"We describe the establishment and current content of the ImmuneCODE™ database, which includes hundreds of millions of T-cell Receptor (TCR) sequences from over 1,400 subjects exposed to or infected with the SARS-CoV-2 virus, as well as over 160,000 high-confidence SARS-CoV-2-specific TCRs.\n\nThis database is made freely available, and the data contained in it can be analyzed online or offline to assist with the global efforts to understand the immune response to the SARS-CoV-2 virus and develop new interventions. Use the \"Open in Analyses\" link now to explore the immune profile of ImmuneCODE subjects in immunoSEQ® Analyzer.\n\nDownload the latest ImmuneCODE datasets here: