1
|
Kristensen NP, Dionisio E, Bentzen AK, Tamhane T, Kemming JS, Nos G, Voss LF, Hansen UK, Lauer GM, Hadrup SR. Simultaneous analysis of pMHC binding and reactivity unveils virus-specific CD8 T cell immunity to a concise epitope set. SCIENCE ADVANCES 2024; 10:eadm8951. [PMID: 38608022 PMCID: PMC11014448 DOI: 10.1126/sciadv.adm8951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
CD8 T cells provide immunity to virus infection through recognition of epitopes presented by peptide major histocompatibility complexes (pMHCs). To establish a concise panel of widely recognized T cell epitopes from common viruses, we combined analysis of TCR down-regulation upon stimulation with epitope-specific enumeration based on barcode-labeled pMHC multimers. We assess CD8 T cell binding and reactivity for 929 previously reported epitopes in the context of 1 of 25 HLA alleles representing 29 viruses. The prevalence and magnitude of CD8 T cell responses were evaluated in 48 donors and reported along with 137 frequently recognized virus epitopes, many of which were underrepresented in the public domain. Eighty-four percent of epitope-specific CD8 T cell populations demonstrated reactivity to peptide stimulation, which was associated with effector and long-term memory phenotypes. Conversely, nonreactive T cell populations were associated primarily with naive phenotypes. Our analysis provides a reference map of epitopes for characterizing CD8 T cell responses toward common human virus infections.
Collapse
Affiliation(s)
- Nikolaj Pagh Kristensen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Edoardo Dionisio
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Amalie Kai Bentzen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Tripti Tamhane
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Janine Sophie Kemming
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Grigorii Nos
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Lasse Frank Voss
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Ulla Kring Hansen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Georg Michael Lauer
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Sine Reker Hadrup
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| |
Collapse
|
2
|
Deering RP, Blumenberg L, Li L, Dhanik A, Jeong S, Pourpe S, Song H, Boucher L, Ragunathan S, Li Y, Zhong M, Kuhnert J, Adler C, Hawkins P, Gupta NT, Moore M, Ni M, Hansen J, Wei Y, Thurston G. Rapid TCR:Epitope Ranker (RAPTER): a primary human T cell reactivity screening assay pairing epitope and TCR at single cell resolution. Sci Rep 2023; 13:8452. [PMID: 37231180 DOI: 10.1038/s41598-023-35710-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 05/22/2023] [Indexed: 05/27/2023] Open
Abstract
Identifying epitopes that T cells respond to is critical for understanding T cell-mediated immunity. Traditional multimer and other single cell assays often require large blood volumes and/or expensive HLA-specific reagents and provide limited phenotypic and functional information. Here, we present the Rapid TCR:Epitope Ranker (RAPTER) assay, a single cell RNA sequencing (scRNA-SEQ) method that uses primary human T cells and antigen presenting cells (APCs) to assess functional T cell reactivity. Using hash-tag oligonucleotide (HTO) coding and T cell activation-induced markers (AIM), RAPTER defines paired epitope specificity and TCR sequence and can include RNA- and protein-level T cell phenotype information. We demonstrate that RAPTER identified specific reactivities to viral and tumor antigens at sensitivities as low as 0.15% of total CD8+ T cells, and deconvoluted low-frequency circulating HPV16-specific T cell clones from a cervical cancer patient. The specificities of TCRs identified by RAPTER for MART1, EBV, and influenza epitopes were functionally confirmed in vitro. In summary, RAPTER identifies low-frequency T cell reactivities using primary cells from low blood volumes, and the resulting paired TCR:ligand information can directly enable immunogenic antigen selection from limited patient samples for vaccine epitope inclusion, antigen-specific TCR tracking, and TCR cloning for further therapeutic development.
Collapse
Affiliation(s)
- Raquel P Deering
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| | - Lili Blumenberg
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Lianjie Li
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Ankur Dhanik
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Se Jeong
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Stephane Pourpe
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Hang Song
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Lauren Boucher
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Shoba Ragunathan
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yanxia Li
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Maggie Zhong
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Jessica Kuhnert
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Christina Adler
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Peter Hawkins
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Namita T Gupta
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Michael Moore
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Johanna Hansen
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Gavin Thurston
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, USA
| |
Collapse
|
3
|
Liu Y, Vieira RMS, Mao L. Simultaneous and Multimodal Antigen-Binding Profiling and Isolation of Rare Cells via Quantitative Ferrohydrodynamic Cell Separation. ACS NANO 2023; 17:94-110. [PMID: 36541668 DOI: 10.1021/acsnano.2c04542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Simultaneous cell profiling and isolation based on cellular antigen-binding capacity plays an important role in understanding and treating diseases. However, fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS) are not able to meet this need, due to their requirement for a large quantity of target cells and the limitation stemming from bimodal separation. Here we report a microfluidic method, termed quantitative ferrohydrodynamic cell separation (qFCS), that achieved multimodal rare cell sorting and simultaneous antigen profiling at a ∼30,000 cell min-1 throughput with a 96.49% recovery rate and a 98.72% purity of recovered cells. qFCS profiles and sorts cells via cellular magnetic content of the magnetically labeled cells, which correlates to cellular antigen-binding capacity. By integrating cellular magnetophoresis and diamagnetophoresis in biocompatible ferrofluids, we demonstrate that the resulting qFCS device can accurately profile and isolate rare cells even when present at ∼1:50,000 target to background cells frequency. We show that the qFCS device could accurately profile and isolate T lymphocytes based on a low-expression CD154 antigen and allow on-device analysis of cells after processing. This method could address the need for simultaneous and multimodal rare cell isolation and profiling in disease diagnostics, prognostics, and treatment.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, The University of Georgia, Athens, Georgia30602, United States
| | - Rafaela Maggioni Simoes Vieira
- J. Crayton Pruitt Family Department of Biomedical Engineering, The University of Florida, Gainesville, Florida32611, United States
| | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, Georgia30602, United States
| |
Collapse
|
4
|
Yu B, Shi Q, Belk JA, Yost KE, Parker KR, Li R, Liu BB, Huang H, Lingwood D, Greenleaf WJ, Davis MM, Satpathy AT, Chang HY. Engineered cell entry links receptor biology with single-cell genomics. Cell 2022; 185:4904-4920.e22. [PMID: 36516854 PMCID: PMC9789208 DOI: 10.1016/j.cell.2022.11.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/31/2022] [Accepted: 11/14/2022] [Indexed: 12/15/2022]
Abstract
Cells communicate with each other via receptor-ligand interactions. Here, we describe lentiviral-mediated cell entry by engineered receptor-ligand interaction (ENTER) to display ligand proteins, deliver payloads, and record receptor specificity. We optimize ENTER to decode interactions between T cell receptor (TCR)-MHC peptides, antibody-antigen, and other receptor-ligand pairs. A viral presentation strategy allows ENTER to capture interactions between B cell receptor and any antigen. We engineer ENTER to deliver genetic payloads to antigen-specific T or B cells to selectively modulate cellular behavior in mixed populations. Single-cell readout of ENTER by RNA sequencing (ENTER-seq) enables multiplexed enumeration of antigen specificities, TCR clonality, cell type, and states of individual T cells. ENTER-seq of CMV-seropositive patient blood samples reveals the viral epitopes that drive effector memory T cell differentiation and inter-clonal vs. intra-clonal phenotypic diversity targeting the same epitope. ENTER technology enables systematic discovery of receptor specificity, linkage to cell fates, and antigen-specific cargo delivery.
Collapse
Affiliation(s)
- Bingfei Yu
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Quanming Shi
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Julia A Belk
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Kevin R Parker
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Betty B Liu
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Huang Huang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Daniel Lingwood
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | | | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
5
|
Klausen U, Grauslund JH, Jørgensen NGD, Ahmad SM, Jonassen M, Weis-Banke SE, Martinenaite E, Pedersen LB, Lisle TL, Gang AO, Enggaard L, Hansen M, Holmström MO, Met Ö, Svane IM, Niemann CU, Pedersen LM, Andersen MH. Anti-PD-L1/PD-L2 therapeutic vaccination in untreated chronic lymphocytic leukemia patients with unmutated IgHV. Front Oncol 2022; 12:1023015. [PMID: 36483037 PMCID: PMC9723164 DOI: 10.3389/fonc.2022.1023015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/28/2022] [Indexed: 09/10/2024] Open
Abstract
Chronic lymphocytic leukemia (CLL) patients with unmutated immunoglobulin heavy chain (IgHV) are at risk of early disease progression compared to patients with mutated IgHV. As a preventive strategy, we treated 19 previously untreated CLL patients with unmutated IgHV in a phase 1/2 trial (clinicaltrials.gov, NCT03939234) exploring the efficacy and toxicity of a therapeutic cancer vaccine containing peptides derived from programmed death ligand 1 (PD-L1) and ligand 2 (PD-L2), hoping to restore immunological control of the disease. According to the International Workshop on Chronic lymphocytic Leukemia (iwCLL) response criteria, no patients obtained a response; however, during follow-up, one patient had complete normalization of the peripheral lymphocyte count and remained in biochemical remission after a follow-up time of 15 months. At the end of treatment, one patient had progressed, and 17 patients had stable disease. During follow-up with a median time of 23.5 months since inclusion, seven patients had progressed, and eight patients had stable disease. The median time to first treatment (TTFT) from diagnosis was 90.3 months with a median follow-up time of 50.1 months. This apparent favorable outcome in TTFT needs to be investigated in a randomized setting, as our population may have been biased. More than 80% of patients obtained vaccine-specific immune responses, confirming the immunogenicity of the vaccine. The vaccine was generally well tolerated with only grade I-II adverse events. Although there were some signs of clinical effects, the vaccine seems to be insufficient as monotherapy in CLL, possibly due to a high tumor burden. The efficacy of the vaccine should preferably be tested in combination with novel targeted therapies or as a consolidating treatment.
Collapse
Affiliation(s)
- Uffe Klausen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nicolai Grønne Dahlager Jørgensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Shamaila Munir Ahmad
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Merete Jonassen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Stine Emilie Weis-Banke
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Evelina Martinenaite
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Lone Bredo Pedersen
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Landkildehus Lisle
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Anne Ortved Gang
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lisbeth Enggaard
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten Hansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Carsten Utoft Niemann
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Garrido-Mesa J, Brown MA. T cell Repertoire Profiling and the Mechanism by which HLA-B27 Causes Ankylosing Spondylitis. Curr Rheumatol Rep 2022; 24:398-410. [PMID: 36197645 PMCID: PMC9666335 DOI: 10.1007/s11926-022-01090-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2022] [Indexed: 11/25/2022]
Abstract
Purpose of Review Ankylosing spondylitis (AS) is strongly associated with the HLA-B27 gene. The canonical function of HLA-B27 is to present antigenic peptides to CD8 lymphocytes, leading to adaptive immune responses. The ‘arthritogenic peptide’ theory as to the mechanism by which HLA-B27 induces ankylosing spondylitis proposes that HLA-B27 presents peptides derived from exogenous sources such as bacteria to CD8 lymphocytes, which subsequently cross-react with antigens at the site of inflammation of the disease, causing inflammation. This review describes findings of studies in AS involving profiling of T cell expansions and discusses future research opportunities based on these findings. Recent Findings Consistent with this theory, there is an expanding body of data showing that expansion of a restricted pool of CD8 lymphocytes is found in most AS patients yet only in a small proportion of healthy HLA-B27 carriers. Summary These exciting findings strongly support the theory that AS is driven by presentation of antigenic peptides to the adaptive immune system by HLA-B27. They point to new potential approaches to identify the exogenous and endogenous antigens involved and to potential therapies for the disease.
Collapse
Affiliation(s)
- Jose Garrido-Mesa
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, England
| | - Matthew A Brown
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, England.
- Genomics England, Charterhouse Square, London, EC1M 6BQ, England.
| |
Collapse
|
7
|
Dobson CS, Reich AN, Gaglione S, Smith BE, Kim EJ, Dong J, Ronsard L, Okonkwo V, Lingwood D, Dougan M, Dougan SK, Birnbaum ME. Antigen identification and high-throughput interaction mapping by reprogramming viral entry. Nat Methods 2022; 19:449-460. [PMID: 35396484 PMCID: PMC9012700 DOI: 10.1038/s41592-022-01436-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 03/01/2022] [Indexed: 01/11/2023]
Abstract
Deciphering immune recognition is critical for understanding a broad range of diseases and for the development of effective vaccines and immunotherapies. Efforts to do so are limited by a lack of technologies capable of simultaneously capturing the complexity of adaptive immunoreceptor repertoires and the landscape of potential antigens. To address this, we present receptor-antigen pairing by targeted retroviruses, which combines viral pseudotyping and molecular engineering approaches to enable one-pot library-on-library interaction screens by displaying antigens on the surface of lentiviruses and encoding their identity in the viral genome. Antigen-specific viral infection of cell lines expressing human T or B cell receptors allows readout of both antigen and receptor identities via single-cell sequencing. The resulting system is modular, scalable and compatible with any cell type. These techniques provide a suite of tools for targeted viral entry, molecular engineering and interaction screens with broad potential applications.
Collapse
Affiliation(s)
- Connor S Dobson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Anna N Reich
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Stephanie Gaglione
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Blake E Smith
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Ellen J Kim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Jiayi Dong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | | | - Vintus Okonkwo
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Michael Dougan
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephanie K Dougan
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael E Birnbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore.
| |
Collapse
|
8
|
Klausen U, Grønne Dahlager Jørgensen N, Grauslund JH, Munir Ahmad S, Gang AO, Martinenaite E, Weis-Banke SE, Breinholt MF, Novotny GW, Kjeldsen JW, Orebo Holmström M, Pedersen LB, Poulsen CB, Hansen PB, Met Ö, Svane IM, Niemann CU, Pedersen LM, Andersen MH. An immunogenic first-in-human immune modulatory vaccine with PD-L1 and PD-L2 peptides is feasible and shows early signs of efficacy in follicular lymphoma. Oncoimmunology 2021; 10:1975889. [PMID: 38283034 PMCID: PMC10813564 DOI: 10.1080/2162402x.2021.1975889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022] Open
Abstract
Cells in the tumor microenvironment of Follicular lymphoma (FL) express checkpoint molecules such as programmed death ligands 1 and 2 (PD-L1 and PD-L2) and are suppressing anti-tumor immune activity. Stimulation of peripheral blood mononuclear cells (PBMC) with PD-L1 (IO103) or PD-L2 (IO120) peptides can activate specific T cells inducing anti-regulatory functions including cytotoxicity against PD-L1/PD-L2-expressing cells. In this study, we vaccinated eight FL patients with PD-L1 and PD-L2 peptides following treatment with standard chemotherapy. Patients experienced grade 1-2 injection site reaction (5/8) and mild flu-like symptoms (6/8). One patient experienced neutropenia and thrombocytopenia during pseudo-progression. Enzyme-linked immunospot detected vaccine-specific immune responses in PBMC from all patients, predominately toward PD-L1. The circulating immune composition was stable during treatment; however, we observed a reduction regulatory T cells, however, not significant. One patient achieved a complete remission during vaccination and two patients had pseudo-progression followed by long-term disease regression. Further examination of these early signs of clinical efficacy of the dual-epitope vaccine in a larger study is warranted.
Collapse
Affiliation(s)
- Uffe Klausen
- Dept. Of Hematology, Herlev Hospital, Herlev, Denmark
- Dept. Of Hematology, Rigshospitalet, Copenhagen, Denmark
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, Copenhagen University, Copenhagen K, Denmark
| | - Nicolai Grønne Dahlager Jørgensen
- Dept. Of Hematology, Herlev Hospital, Herlev, Denmark
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
| | - Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, Copenhagen University, Copenhagen K, Denmark
| | - Shamaila Munir Ahmad
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
| | | | - Evelina Martinenaite
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
| | - Stine Emilie Weis-Banke
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
| | | | - Guy Wayne Novotny
- Dept. Of Hematology, Herlev Hospital, Herlev, Denmark
- Dept. Of Pathology, Herlev Hospital, Herlev, Denmark
| | - Julie Westerlin Kjeldsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, Copenhagen University, Copenhagen K, Denmark
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, Copenhagen University, Copenhagen K, Denmark
| | | | | | - Per Boye Hansen
- Dept. Of Hematology, Zealand University Hospital, Roskilde, Rosklide, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, Copenhagen University, Copenhagen K, Denmark
| | - Carsten Utoft Niemann
- Dept. Of Hematology, Rigshospitalet, Copenhagen, Denmark
- Institute for Immunology and Microbiology, Copenhagen University, Copenhagen K, Denmark
| | - Lars Møller Pedersen
- Dept. Of Hematology, Herlev Hospital, Herlev, Denmark
- Dept. Of Hematology, Rigshospitalet, Copenhagen, Denmark
- Dept. Of Hematology, Zealand University Hospital, Roskilde, Rosklide, Denmark
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology Herlev Hospital, Herlev, Denmark
- Institute for Immunology and Microbiology, Copenhagen University, Copenhagen K, Denmark
| |
Collapse
|
9
|
Michelo CM, Dalel JA, Hayes P, Fernandez N, Fiore-Gartland A, Kilembe W, Tang J, Streatfield C, Gilmour J, Hunter E. Comprehensive epitope mapping using polyclonally expanded human CD8 T cells and a two-step ELISpot assay for testing large peptide libraries. J Immunol Methods 2021; 491:112970. [PMID: 33529681 PMCID: PMC8008507 DOI: 10.1016/j.jim.2021.112970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/30/2020] [Accepted: 01/18/2021] [Indexed: 02/01/2023]
Abstract
The genetic diversity of circulating HIV-1 strains poses a major barrier to the design, development and evaluation of HIV-1 vaccines. The assessment of both vaccine- and natural infection-elicited T cell responses is commonly done with multivalent peptides that are designed to maximally capture the diversity of potential T cell epitopes (PTEs) observed in natural circulating sequences. However, depending on the sequence diversity of viral subtypes and number of the HIV immunogens under investigation, PTE estimates, including HLA-guided computational methods, can easily generate enormous peptide libraries. Evaluation of T cell epitope specificity using such extensive peptide libraries is usually limited by sample availability, even for high-throughput and robust epitope mapping techniques like ELISpot assays. Here we describe a novel, two-step protocol for in-vitro polyclonal expansion of CD8 T cells from a single vial of frozen PBMC, which facilitated the screening 441 HIV-1 Gag peptides for immune responses among 32 HIV-1 positive subjects and 40 HIV-1 negative subjects for peptide qualification. Using a pooled-peptide mapping strategy, epitopes were mapped in two sequential ELISpot assays; the first ELISpot screened 33 large peptide pools using CD8 T cells expanded for 7 days, while the second step tested pool-matrix peptides to identify individual peptides using CD8 T cells expanded for 10 days. This comprehensive epitope screening established the breadth and magnitude of HIV-1 Gag-specific CD8 T cells and further revealed the extent of immune responses to variable/polymorphic epitopes.
Collapse
Affiliation(s)
- Clive M Michelo
- Zambia Emory HIV Research Project, B22/737 Mwembelelo, Emmasdale, Lusaka, Zambia
| | - Jama A Dalel
- Human Immunology Laboratory, International AIDS Vaccine Initiative, Imperial College London, London, United Kingdom
| | - Peter Hayes
- Human Immunology Laboratory, International AIDS Vaccine Initiative, Imperial College London, London, United Kingdom
| | - Natalia Fernandez
- Human Immunology Laboratory, International AIDS Vaccine Initiative, Imperial College London, London, United Kingdom
| | - Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - William Kilembe
- Zambia Emory HIV Research Project, B22/737 Mwembelelo, Emmasdale, Lusaka, Zambia
| | - Jianming Tang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Claire Streatfield
- Human Immunology Laboratory, International AIDS Vaccine Initiative, Imperial College London, London, United Kingdom
| | - Jill Gilmour
- Human Immunology Laboratory, International AIDS Vaccine Initiative, Imperial College London, London, United Kingdom
| | - Eric Hunter
- Zambia Emory HIV Research Project, B22/737 Mwembelelo, Emmasdale, Lusaka, Zambia; Emory Vaccine Center, Yerkes National Primate Research Center, 954 Gatewood Road NE, Atlanta, GA 30329, USA.
| |
Collapse
|
10
|
Jørgensen NG, Kaae J, Grauslund JH, Met Ö, Nielsen SL, Pedersen AW, Svane IM, Ehrnrooth E, Andersen MH, Zachariae C, Skov L. Vaccination against PD-L1 with IO103 a Novel Immune Modulatory Vaccine in Basal Cell Carcinoma: A Phase IIa Study. Cancers (Basel) 2021; 13:cancers13040911. [PMID: 33671555 PMCID: PMC7926323 DOI: 10.3390/cancers13040911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 01/24/2023] Open
Abstract
Simple Summary Basal cell carcinoma is the most common skin cancer and new treatments for patients with widespread and numerous tumors are lacking. In a previous study treating patients with multiple myeloma with a peptide vaccine called IO103 against an immune checkpoint molecule called programmed death ligand 1, two cases of basal cell carcinoma regressed. The aim of the present study was to assess the effect of vaccination with IO103 in ten patients with basal cell carcinoma. Patients were vaccinated with Montanide as adjuvant up to nine times during six months. Regression in tumor size of at least 30% was seen in five of 18 tumors, two of which showed complete regression. Vaccinations resulted in immune responses against the vaccine in blood samples from nine of ten patients and in skin samples from five of nine patients. The findings suggest that the vaccine may be effective against some basal cell carcinomas. Abstract Antitumor activity of immune checkpoint blocking antibodies against programmed death 1 (PD-1) in basal cell carcinoma (BCC) has been described. IO103 is a peptide vaccine against the major PD-1 ligand PD-L1. A phase IIa study of vaccination with IO103 and Montanide adjuvant was conducted in patients with resectable BCC (NCT03714529). Vaccinations were given six times every 2 weeks (q2w), followed by three vaccines q4w in responders. Primary endpoints were clinical responses of target tumors, change in target tumor size and immune responses to the vaccine. Secondary endpoint was safety. One tumor per patient was designated target tumor and biopsied twice during the course of vaccination. Synchronous non-target BCCs were not biopsied during vaccinations. Ten patients were vaccinated (six patients received six vaccinations and four patients received nine vaccinations). A partial response (PR) was seen in two target tumors. Two complete responses (CR) and one PR were observed in eight non-target tumors in four patients. No tumors progressed. Related adverse events were grade 1 and reversible. Immune responses against IO103 were induced in blood samples from nine of ten and skin-infiltrating lymphocytes from five of the nine patients. The regressions seen in non-target tumors suggest that IO103 may be effective against a subtype of BCC.
Collapse
Affiliation(s)
- Nicolai Grønne Jørgensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark; (J.H.G.); (Ö.M.); (I.M.S.); (M.H.A.)
- Department of Hematology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark
- Correspondence:
| | - Jeanette Kaae
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark; (J.K.); (C.Z.); (L.S.)
| | - Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark; (J.H.G.); (Ö.M.); (I.M.S.); (M.H.A.)
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark; (J.H.G.); (Ö.M.); (I.M.S.); (M.H.A.)
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Signe Ledou Nielsen
- Department of Pathology, Herlev and Gentofte Hospital, 2730 Herlev, Denmark;
| | | | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark; (J.H.G.); (Ö.M.); (I.M.S.); (M.H.A.)
| | - Eva Ehrnrooth
- IO Biotech ApS, 2200 Copenhagen, Denmark; (A.W.P.); (E.E.)
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark; (J.H.G.); (Ö.M.); (I.M.S.); (M.H.A.)
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
- IO Biotech ApS, 2200 Copenhagen, Denmark; (A.W.P.); (E.E.)
| | - Claus Zachariae
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark; (J.K.); (C.Z.); (L.S.)
| | - Lone Skov
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark; (J.K.); (C.Z.); (L.S.)
| |
Collapse
|
11
|
Køllgaard T, Klausen TW, Idorn M, Holmgaard RB, Straten PT, Andersen MH. Association of a functional Indoleamine 2,3-dioxygenase 2 genotype with specific immune responses. Oncoimmunology 2021; 1:441-447. [PMID: 22754762 PMCID: PMC3382899 DOI: 10.4161/onci.19654] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Two frequent single-nucleotide-polymorphisms (SNPs) are present in the indoleamine 2,3-dioxygenase 2 (IDO2) gene that influence its enzymatic activity. Thus, one SNP (R248W) is associated with a reduction in IDO2 catalytic activity, whereas the other SNP (Y359stop) generates a premature stop codon abolishing activity completely. In the present study, we describe the presence of a specific cellular immune response in the periphery which correlated with the functional status of the IDO2 protein. Hence, the induction of IDO2-specific T cells in peripheral blood requires the presence of a functional IDO2 protein and, consequently, is restricted to individuals that are not homozygous for the stop codon. Furthermore, we detected stronger T-cell responses in donors with the homozygous Y wild type at position 359 when compared with the heterozygous genotype. Interestingly, we found a higher number of immune responses against IDO2 in patients homozygous for the 248W giving reduction in IDO2 activity compared with the 248R. Hence, spontaneous immune responses against IDO2 seem to be correlated with reduced enzymatic activity of IDO2. The patient IDO2 genotype may well influence the outcome of IDO2-based anti-cancer vaccination.
Collapse
Affiliation(s)
- Tania Køllgaard
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital; Herlev, Denmark
| | | | | | | | | | | |
Collapse
|
12
|
Munir S, Frøsig TM, Hansen M, Svane IM, Andersen MH. Characterization of T-cell responses against IκBα in cancer patients. Oncoimmunology 2021; 1:1290-1296. [PMID: 23243592 PMCID: PMC3518501 DOI: 10.4161/onci.21625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The nuclear factor κ light chain enhancer of activated B cells (NFκB) is constitutively active in most cancers, controlling multiple cellular processes including proliferation, invasion and resistance to therapy. NFκB is primarily regulated through the association with inhibitory proteins that are known as inhibitors of NFκB (IκBs). Increased NFκB activity in tumor cells has been correlated with decrease stability of IκB proteins, in particular IκBα. In responso to a large number of stimuli, IκB proteins are degraded by the proteasome. Cytotoxic T lymphocytes (CTLs) recognize HLA-restricted antigenic peptides that are generated by proteasomal degradation in target cells. In the present study, we demonstrate the presence of naturally occurring IκBα -specific T cells in the peripheral blood of patients suffering from several unrelated tumor types, i.e., breast cancer, malignant melanoma and renal cell carcinoma, but not of healthy controls. Furthermore, we show that such IBα-specific T cells are granzyme B-releasing, cytotoxic cells. Hence, the increased proteasomal degradation of IκBα in cancer induces IκBα-specific CTLs.
Collapse
Affiliation(s)
- Shamaila Munir
- Center for Cancer Immune Therapy (CCIT); Department of Hematology and Oncology; Copenhagen University Hospital; Herlev, Denmark
| | | | | | | | | |
Collapse
|
13
|
Bousbaine D, Ploegh HL. Antigen discovery tools for adaptive immune receptor repertoire research. CURRENT OPINION IN SYSTEMS BIOLOGY 2020; 24:64-70. [PMID: 33195881 PMCID: PMC7665270 DOI: 10.1016/j.coisb.2020.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The adaptive immune system has evolved to recognize with incredible precision a large diversity of molecules. Innovations in high-throughput sequencing and bioinformatics have accelerated large-scale immune repertoire analyses and given us important insights into the behavior of the adaptive immune system. However, establishing a connection between receptor sequence and its antigen-specificity remains a challenge despite its central role in determining T and B cell fate. We discuss recent large-scale antigen discovery technologies which can be combined with adaptive immune receptor repertoire (AIRR) studies. We highlight important discoveries made using repertoire analyses in the field of host-microbe interactions.
Collapse
Affiliation(s)
- Djenet Bousbaine
- Department of Bioengineering and ChEM-H, Stanford University, Stanford CA, USA
| | - Hidde L. Ploegh
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston MA, USA
| |
Collapse
|
14
|
Jørgensen NG, Klausen U, Grauslund JH, Helleberg C, Aagaard TG, Do TH, Ahmad SM, Olsen LR, Klausen TW, Breinholt MF, Hansen M, Martinenaite E, Met Ö, Svane IM, Knudsen LM, Andersen MH. Peptide Vaccination Against PD-L1 With IO103 a Novel Immune Modulatory Vaccine in Multiple Myeloma: A Phase I First-in-Human Trial. Front Immunol 2020; 11:595035. [PMID: 33240282 PMCID: PMC7680803 DOI: 10.3389/fimmu.2020.595035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Background Immune checkpoint blockade with monoclonal antibodies targeting programmed death 1 (PD-1) and its ligand PD-L1 has played a major role in the rise of cancer immune therapy. We have identified naturally occurring self-reactive T cells specific to PD-L1 in both healthy donors and cancer patients. Stimulation with a PD-L1 peptide (IO103), activates these cells to exhibit inflammatory and anti-regulatory functions that include cytotoxicity against PD-L1-expressing target cells. This prompted the initiation of the present first-in-human study of vaccination with IO103, registered at clinicaltrials.org (NCT03042793). Methods Ten patients with multiple myeloma who were up to 6 months after high dose chemotherapy with autologous stem cell support, were enrolled. Subcutaneous vaccinations with IO103 with the adjuvant Montanide ISA 51 was given up to fifteen times during 1 year. Safety was assessed by the common toxicity criteria for adverse events (CTCAE). Immunogenicity of the vaccine was evaluated using IFNγ enzyme linked immunospot and intracellular cytokine staining on blood and skin infiltrating lymphocytes from sites of delayed-type hypersensitivity. The clinical course was described. Results All adverse reactions to the PD-L1 vaccine were below CTCAE grade 3, and most were grade 1-2 injection site reactions. The total rate of adverse events was as expected for the population. All patients exhibited peptide specific immune responses in peripheral blood mononuclear cells and in skin-infiltrating lymphocytes after a delayed-type hypersensitivity test. The clinical course was as expected for the population. Three of 10 patients had improvements of responses which coincided with the vaccinations. Conclusion Vaccination against PD-L1 was associated with low toxicity and high immunogenicity. This study has prompted the initiation of later phase trials to assess the vaccines efficacy. Clinical Trial Registration clinicaltrials.org, identifier NCT03042793.
Collapse
Affiliation(s)
- Nicolai Grønne Jørgensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.,Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Uffe Klausen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.,Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Carsten Helleberg
- Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Trung Hieu Do
- Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Shamaila Munir Ahmad
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | | | - Morten Hansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Evelina Martinenaite
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Abstract
T cells respond to threats in an antigen-specific manner using T cell receptors (TCRs) that recognize short peptide antigens presented on major histocompatibility complex (MHC) proteins. The TCR-peptide-MHC interaction mediated between a T cell and its target cell dictates its function and thereby influences its role in disease. A lack of approaches for antigen discovery has limited the fundamental understanding of the antigenic landscape of the overall T cell response. Recent advances in high-throughput sequencing, mass cytometry, microfluidics and computational biology have led to a surge in approaches to address the challenge of T cell antigen discovery. Here, we summarize the scope of this challenge, discuss in depth the recent exciting work and highlight the outstanding questions and remaining technical hurdles in this field.
Collapse
|
16
|
Draxler DF, Daglas M, Fernando A, Hanafi G, McCutcheon F, Ho H, Galle A, Gregory J, Larsson P, Keragala C, Wright DK, Tavancheh E, Au AE, Niego B, Wilson K, Plebanski M, Sashindranath M, Medcalf RL. Tranexamic acid modulates the cellular immune profile after traumatic brain injury in mice without hyperfibrinolysis. J Thromb Haemost 2019; 17:2174-2187. [PMID: 31393041 DOI: 10.1111/jth.14603] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 07/30/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is known to promote immunosuppression, making patients more susceptible to infection, yet potentially exerting protective effects by inhibiting central nervous system (CNS) reactivity. Plasmin, the effector protease of the fibrinolytic system, is now recognized for its involvement in modulating immune function. OBJECTIVE To evaluate the effects of plasmin and tranexamic acid (TXA) on the immune response in wild-type and plasminogen-deficient (plg-/- ) mice subjected to TBI. METHODS Leukocyte subsets in lymph nodes and the brain in mice post TBI were evaluated by flow cytometry and in blood with a hemocytometer. Immune responsiveness to CNS antigens was determined by Enzyme-linked Immunosorbent Spot (ELISpot) assay. Fibrinolysis was determined by thromboelastography and measuring D-dimer and plasmin-antiplasmin complex levels. RESULTS Plg-/- mice, but not plg+/+ mice displayed increases in both the number and activation of various antigen-presenting cells and T cells in the cLN 1 week post TBI. Wild-type mice treated with TXA also displayed increased cellularity of the cLN 1 week post TBI together with increases in innate and adaptive immune cells. These changes occurred despite the absence of systemic hyperfibrinolysis or coagulopathy in this model of TBI. Importantly, neither plg deficiency nor TXA treatment enhanced the autoreactivity within the CNS. CONCLUSION In the absence of systemic hyperfibrinolysis, plasmin deficiency or blockade with TXA increases migration and proliferation of conventional dendritic cells (cDCs) and various antigen-presenting cells and T cells in the draining cervical lymph node (cLN) post TBI. Tranexamic acid might also be clinically beneficial in modulating the inflammatory and immune response after TBI, but without promoting CNS autoreactivity.
Collapse
Affiliation(s)
- Dominik F Draxler
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Maria Daglas
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Anushka Fernando
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Gryselda Hanafi
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Fiona McCutcheon
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Heidi Ho
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Adam Galle
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Julia Gregory
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Pia Larsson
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Charithani Keragala
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Elnaz Tavancheh
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Amanda E Au
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Be'eri Niego
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Kirsty Wilson
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Maithili Sashindranath
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Robert L Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
17
|
Early Diagnosis of Pathogen Infection by Cell-Based Activation Immunoassay. Cells 2019; 8:cells8090952. [PMID: 31443439 PMCID: PMC6769711 DOI: 10.3390/cells8090952] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 12/12/2022] Open
Abstract
Diagnostic identification of pathogens is usually accomplished by isolation of the pathogen or its substances, and should correlate with the time and site of infection. Alternatively, immunoassays such as enzyme-linked immunosorbent assay (ELISA) tests for quantification of serum antibodies are expedient and are usually employed for retrospective diagnostic of a particular infective agent. Here, the potential of cell-based immunoassays for early pathogen detection was evaluated by quantification of specific, antigen-activated, low-frequency IFNγ-secreting cells in mouse spleens following infection with various pathogens. Using enzyme-linked immunospot (ELISPOT) assays, specific responses were observed within 3–6 days following infection with F. tularensis, B. anthracis, Y. pestis, or Influenza virus. Blood samples collected from F. tularensis-infected mice revealed the presence of IFNγ-producing activated cells within one week post infection. When non-human primates were infected with B. anthracis, cellular response was observed in peripheral blood samples as early as five days post infection, 3–5 days earlier than serum antibodies. Finally, the expression pattern of genes in splenocytes of F. tularensis-infected mice was inspected by a transcriptomic approach, enabling the identification of potential host targets for the future development of genetic-based cellular immunoassays. Altogether, the data demonstrate the potential of cell-based immunoassays for early pathogen detection.
Collapse
|
18
|
Simoni Y, Chng MHY, Li S, Fehlings M, Newell EW. Mass cytometry: a powerful tool for dissecting the immune landscape. Curr Opin Immunol 2018; 51:187-196. [PMID: 29655022 DOI: 10.1016/j.coi.2018.03.023] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/25/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
Abstract
Advancement in methodologies for single cell analysis has historically been a major driver of progress in immunology. Currently, high dimensional flow cytometry, mass cytometry and various forms of single cell sequencing-based analysis methods are being widely adopted to expose the staggering heterogeneity of immune cells in many contexts. Here, we focus on mass cytometry, a form of flow cytometry that allows for simultaneous interrogation of more than 40 different marker molecules, including cytokines and transcription factors, without the need for spectral compensation. We argue that mass cytometry occupies an important niche within the landscape of single-cell analysis platforms that enables the efficient and in-depth study of diverse immune cell subsets with an ability to zoom-in on myeloid and lymphoid compartments in various tissues in health and disease. We further discuss the unique features of mass cytometry that are favorable for combining multiplex peptide-MHC multimer technology and phenotypic characterization of antigen specific T cells. By referring to recent studies revealing the complexities of tumor immune infiltrates, we highlight the particular importance of this technology for studying cancer in the context of cancer immunotherapy. Finally, we provide thoughts on current technical limitations and how we imagine these being overcome.
Collapse
Affiliation(s)
- Yannick Simoni
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Melissa Hui Yen Chng
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Shamin Li
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | | | - Evan W Newell
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore.
| |
Collapse
|
19
|
Lam JKP, Hui KF, Ning RJ, Xu XQ, Chan KH, Chiang AKS. Emergence of CD4+ and CD8+ Polyfunctional T Cell Responses Against Immunodominant Lytic and Latent EBV Antigens in Children With Primary EBV Infection. Front Microbiol 2018; 9:416. [PMID: 29599759 PMCID: PMC5863510 DOI: 10.3389/fmicb.2018.00416] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/21/2018] [Indexed: 11/13/2022] Open
Abstract
Long term carriers were shown to generate robust polyfunctional T cell (PFC) responses against lytic and latent antigens of Epstein-Barr virus (EBV). However, the time of emergence of PFC responses against EBV antigens, pattern of immunodominance and difference between CD4+ and CD8+ T cell responses during various stages of EBV infection are not clearly understood. A longitudinal study was performed to assess the development of antigen-specific PFC responses in children diagnosed to have primary symptomatic (infectious mononucleosis [IM]) and asymptomatic (AS) EBV infection. Evaluation of IFN-γ secreting CD8+ T cell responses upon stimulation by HLA class I-specific peptides of EBV lytic and latent proteins by ELISPOT assay followed by assessment of CD4+ and CD8+ PFC responses upon stimulation by a panel of overlapping EBV peptides for co-expression of IFN-γ, TNF-α, IL-2, perforin and CD107a by flow cytometry were performed. Cytotoxicity of T cells against autologous lymphoblastoid cell lines (LCLs) as well as EBV loads in PBMC and plasma were also determined. Both IM and AS patients had elevated PBMC and plasma viral loads which declined steadily during a 12-month period from the time of diagnosis whilst decrease in the magnitude of CD8+ T cell responses toward EBV lytic peptides in contrast to increase toward latent peptides was shown with no significant difference between those of IM and AS patients. Both lytic and latent antigen-specific CD4+ and CD8+ T cells demonstrated polyfunctionality (defined as greater or equal to three functions) concurrent with enhanced cytotoxicity against autologous LCLs and steady decrease in plasma and PBMC viral loads over time. Immunodominant peptides derived from BZLF1, BRLF1, BMLF1 and EBNA3A-C proteins induced the highest proportion of CD8+ as well as CD4+ PFC responses. Diverse functional subtypes of both CD4+ and CD8+ PFCs were shown to emerge at 6–12 months. In conclusion, EBV antigen-specific CD4+ and CD8+ PFC responses emerge during the first year of primary EBV infection, with greatest responses toward immunodominant epitopes in both lytic and latent proteins, correlating to steady decline in PBMC and plasma viral loads.
Collapse
Affiliation(s)
- Janice K P Lam
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - K F Hui
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Raymond J Ning
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - X Q Xu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - K H Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Alan K S Chiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
20
|
Baumgaertner P, Ioannidou K, Speiser DE. Immune Monitoring of Blood and Tumor Microenvironment. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
21
|
Möbs C, Schmidt T. Research Techniques Made Simple: Monitoring of T-Cell Subsets using the ELISPOT Assay. J Invest Dermatol 2017; 136:e55-e59. [PMID: 27212652 DOI: 10.1016/j.jid.2016.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The enzyme-linked immunospot (ELISPOT) assay allows characterization of single-cell immune responses through detection of secreted analytes. Although ELISPOT analysis shares similarities with ELISA, it has some essential differences. In general, the ELISPOT assay uses antibodies to capture and detect analytes of interest released by activated immune cells. Released analytes form specific antibody-antigen complexes and are visualized as spots of enzyme-substrate precipitates. These spots indicate both how many cells secrete the respective analyte and how much analyte is produced per individual cell. Initially developed for the detection of antibody-secreting cells, ELISPOT assays are now frequently performed both in the context of clinical diagnostics and in research on T-cell responses, in particular antigen-specific T-cell subpopulations, as related to allergy, cancer, infections, or autoimmune diseases. The one spot-one cell principle allows sensitive detection of specific and rare immune cell subsets. Here we present general principles, applications, and recent modifications of the ELISPOT technique.
Collapse
Affiliation(s)
- Christian Möbs
- Department of Dermatology and Allergology, Philipps University Marburg, Marburg, Germany.
| | - Thomas Schmidt
- Department of Dermatology and Allergology, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
22
|
Jørgensen NG, Ahmad SM, Abildgaard N, Straten PT, Svane IM, Andersen MH, Knudsen LM. Peptide vaccination against multiple myeloma using peptides derived from anti-apoptotic proteins: a phase I trial. Stem Cell Investig 2016; 3:95. [PMID: 28078275 PMCID: PMC5222710 DOI: 10.21037/sci.2016.11.09] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 11/07/2016] [Indexed: 11/06/2022]
Abstract
The B-cell lymphoma-2 (Bcl-2) family of proteins play a crucial role in multiple myeloma (MM), contributing to lacking apoptosis which is a hallmark of the disease. This makes the Bcl-2 proteins interesting targets for therapeutic peptide vaccination. We report a phase I trial of therapeutic vaccination with peptides from the proteins Bcl-2, Bcl-XL and Mcl-1 in patients with relapsed MM. Vaccines were given concomitant with bortezomib. Out of 7 enrolled patients, 4 received the full course of 8 vaccinations. The remaining 3 patients received fewer vaccinations due to progression, clinical decision of lacking effect and development of hypercalcemia, respectively. There were no signs of toxicity other than what was to be expected from bortezomib. Immune responses to the peptides were seen in all 6 patients receiving more than 2 vaccinations. Three patients had increased immune responses after vaccination. Vaccination against Bcl-2 was well tolerated and was able to induce immune responses in patients with relapsed MM.
Collapse
Affiliation(s)
- Nicolai Grønne Jørgensen
- Center for Cancer Immunotherapy (CCIT), Department of Hematology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Shamaila Munir Ahmad
- Center for Cancer Immunotherapy (CCIT), Department of Hematology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Niels Abildgaard
- Department of Hematology, University Hospital Odense, Odense, Denmark
| | - Per Thor Straten
- Center for Cancer Immunotherapy (CCIT), Department of Hematology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immunotherapy (CCIT), Department of Hematology, Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immunotherapy (CCIT), Department of Hematology, Herlev and Gentofte Hospital, Herlev, Denmark
| | | |
Collapse
|
23
|
Bentzen AK, Marquard AM, Lyngaa R, Saini SK, Ramskov S, Donia M, Such L, Furness AJS, McGranahan N, Rosenthal R, Straten PT, Szallasi Z, Svane IM, Swanton C, Quezada SA, Jakobsen SN, Eklund AC, Hadrup SR. Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes. Nat Biotechnol 2016; 34:1037-1045. [PMID: 27571370 DOI: 10.1038/nbt.3662] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/04/2016] [Indexed: 01/17/2023]
Abstract
Identification of the peptides recognized by individual T cells is important for understanding and treating immune-related diseases. Current cytometry-based approaches are limited to the simultaneous screening of 10-100 distinct T-cell specificities in one sample. Here we use peptide-major histocompatibility complex (MHC) multimers labeled with individual DNA barcodes to screen >1,000 peptide specificities in a single sample, and detect low-frequency CD8 T cells specific for virus- or cancer-restricted antigens. When analyzing T-cell recognition of shared melanoma antigens before and after adoptive cell therapy in melanoma patients, we observe a greater number of melanoma-specific T-cell populations compared with cytometry-based approaches. Furthermore, we detect neoepitope-specific T cells in tumor-infiltrating lymphocytes and peripheral blood from patients with non-small cell lung cancer. Barcode-labeled pMHC multimers enable the combination of functional T-cell analysis with large-scale epitope recognition profiling for the characterization of T-cell recognition in various diseases, including in small clinical samples.
Collapse
Affiliation(s)
- Amalie Kai Bentzen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Andrea Marion Marquard
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Rikke Lyngaa
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Sunil Kumar Saini
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Sofie Ramskov
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Lina Such
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Andrew J S Furness
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Cancer Immunology Unit, UCL Cancer Institute, University College London, London, UK
| | - Nicholas McGranahan
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Rachel Rosenthal
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Per Thor Straten
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Zoltan Szallasi
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Charles Swanton
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Sergio A Quezada
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Cancer Immunology Unit, UCL Cancer Institute, University College London, London, UK
| | - Søren Nyboe Jakobsen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
- Immudex, Copenhagen, Denmark
| | - Aron Charles Eklund
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| |
Collapse
|
24
|
Abstract
Membranes are widely used as protein blotting matrices for a large variety of research applications including western blotting and enzyme-linked immunospot assay (ELISPOT). The largest advantage of using membranes versus solid plastic support is the porosity of membranes allowing for immobilization of high concentrations of proteins and antibodies which, in turn, increases the sensitivity of detection. Similar to plastic surfaces, polyvinylidene difluoride (PVDF) and nitrocellulose membranes create good microenvironment for live cells cultured in vitro and do not interfere with cellular physiology. It appears that PVDF-backed microplates are a golden standard for ELISPOT assays: such plates are inexpensive, easy to use and after assay development, membranes can be removed from the plates and archived. Given the convenience and reliability of membrane microplates, they are widely used in ELISPOT assays for basic research and clinical trials. The ELISPOT assay is an antibody "sandwich" technique aimed at trapping cell-secreted molecules between capture and detection antibodies, followed by either chromogenic enzymatic or fluorescence detection. This review covers the principles of the ELISPOT assay on membrane microplates including single-color and two-color detection techniques with the emphasis on assay design, choosing membrane microplates, and troubleshooting protocols.
Collapse
|
25
|
Normal Distribution of CD8+ T-Cell-Derived ELISPOT Counts within Replicates Justifies the Reliance on Parametric Statistics for Identifying Positive Responses. Cells 2015; 4:96-111. [PMID: 25738924 PMCID: PMC4381212 DOI: 10.3390/cells4010096] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 11/16/2022] Open
Abstract
Accurate assessment of positive ELISPOT responses for low frequencies of antigen-specific T-cells is controversial. In particular, it is still unknown whether ELISPOT counts within replicate wells follow a theoretical distribution function, and thus whether high power parametric statistics can be used to discriminate between positive and negative wells. We studied experimental distributions of spot counts for up to 120 replicate wells of IFN-γ production by CD8+ T-cell responding to EBV LMP2A (426 - 434) peptide in human PBMC. The cells were tested in serial dilutions covering a wide range of average spot counts per condition, from just a few to hundreds of spots per well. Statistical analysis of the data using diagnostic Q-Q plots and the Shapiro-Wilk normality test showed that in the entire dynamic range of ELISPOT spot counts within replicate wells followed a normal distribution. This result implies that the Student t-Test and ANOVA are suited to identify positive responses. We also show experimentally that borderline responses can be reliably detected by involving more replicate wells, plating higher numbers of PBMC, addition of IL-7, or a combination of these. Furthermore, we have experimentally verified that the number of replicates needed for detection of weak responses can be calculated using parametric statistics.
Collapse
|
26
|
Larsen SK, Ahmad SM, Idorn M, Met Ö, Martinenaite E, Svane IM, Straten PT, Andersen MH. Spontaneous presence of FOXO3-specific T cells in cancer patients. Oncoimmunology 2014; 3:e953411. [PMID: 25960934 DOI: 10.4161/21624011.2014.953411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/02/2014] [Indexed: 02/07/2023] Open
Abstract
In the present study, we describe forkhead box O3 (FOXO3)-specific, cytotoxic CD8+ T cells existent among peripheral-blood mononuclear cells (PBMCs) of cancer patients. FOXO3 immunogenicity appears specific, as we did not detect reactivity toward FOXO3 among T cells in healthy individuals. FOXO3 may naturally serve as a target antigen for tumor-reactive T cells as it is frequently over-expressed in cancer cells. In addition, expression of FOXO3 plays a critical role in immunosuppression mediated by tumor-associated dendritic cells (TADCs). Indeed, FOXO3-specific cytotoxic T lymphocytes (CTLs) were able to specifically recognize and kill both FOXO3-expressing cancer cells as well as dendritic cells. Thus, FOXO3 was processed and presented by HLA-A2 on the cell surface of both immune cells and cancer cells. As FOXO3 programs TADCs to become tolerogenic, FOXO3 signaling thereby comprises a significant immunosuppressive mechanism, such that FOXO3 targeting by means of specific T cells is an attractive clinical therapy to boost anticancer immunity. In addition, the natural occurrence of FOXO3-specific CTLs in the periphery suggests that these T cells hold a function in the complex network of immune regulation in cancer patients.
Collapse
Key Words
- APC, antigen presenting cell
- CTL
- CTL, cytotoxic T lymphocyte
- CTLA4, cytotoxic T-lymphocyte associated protein 4
- DC, dendritic cell
- FOXO3
- FOXO3, forkhead box O3
- IDO, indoleamine-2,3-dioxygenase
- PBMC, peripheral blood mononuclear cell
- TADC, tumor-associated DCs
- TGFβ, tumor growth factor β
- TNFα, tumor necrosis factor α
- Tregs, regulatory T cell
- antigens
- immune regulation
- tumor-associated dendritic cells
Collapse
Affiliation(s)
- Stine Kiaer Larsen
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark ; These authors contributed equally to this work
| | - Shamaila Munir Ahmad
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark ; These authors contributed equally to this work
| | - Manja Idorn
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Özcan Met
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Evelina Martinenaite
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Per Thor Straten
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital ; Herlev ; Herlev, Denmark
| |
Collapse
|
27
|
A novel oil-in-water emulsion as a potential adjuvant for influenza vaccine: Development, characterization, stability and in vivo evaluation. Int J Pharm 2014; 468:187-95. [DOI: 10.1016/j.ijpharm.2014.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/15/2014] [Accepted: 04/02/2014] [Indexed: 11/21/2022]
|
28
|
Ridolfi R, Ridolfi L, Petrini M, Fiammenghi L, Riccobon A. Dendritic cell vaccination and immunostimulation in advanced melanoma. Expert Rev Vaccines 2014; 2:825-33. [PMID: 14711365 DOI: 10.1586/14760584.2.6.825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The most recent advances in immunology bear witness to the fact that tumors, in particular melanoma, escape recognition by the host's immune system and can locally inactivate its effectors, T-cells and antigen presenting cells. There is, however, preclinical evidence that the immune system, opportunely stimulated, is capable of recognizing and killing tumor cells. It has been verified that the activation of autologous dendritic cells, derived from peripheral blood and pulsed with tumor antigens, results in the specific stimulation of T-cells against the tumor. Preliminary data from dendritic cell vaccination trials, mainly of advanced melanoma, show unequivocal evidence of immunization and of the first clinical responses. Many questions remain to be answered before more effective and widespread use of this type of vaccination is possible, especially in the early stages of the disease.
Collapse
Affiliation(s)
- Ruggero Ridolfi
- Dept. of Medical Oncology, Pierantoni Hospital, Via Forlanini 34, 47100 Forlì, Italy.
| | | | | | | | | |
Collapse
|
29
|
Iversen TZ, Engell-Noerregaard L, Ellebaek E, Andersen R, Larsen SK, Bjoern J, Zeyher C, Gouttefangeas C, Thomsen BM, Holm B, Thor Straten P, Mellemgaard A, Andersen MH, Svane IM. Long-lasting disease stabilization in the absence of toxicity in metastatic lung cancer patients vaccinated with an epitope derived from indoleamine 2,3 dioxygenase. Clin Cancer Res 2013; 20:221-32. [PMID: 24218513 DOI: 10.1158/1078-0432.ccr-13-1560] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate targeting of indoleamine 2,3 dioxygenase (IDO) enzyme using a synthetic peptide vaccine administered to patients with metastatic non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN In a clinical phase I study, we treated 15 HLA-A2-positive patients with stage III-IV NSCLC in disease stabilization after standard chemotherapy. Patients were treated with imiquimod ointment and subcutaneous vaccinations (100 μg IDO5 peptide, sequence ALLEIASCL, formulated in 900 μL Montanide). Primary endpoint was toxicity. Clinical benefit and immunity were assessed as secondary endpoints. RESULTS No severe toxicity was observed. One patient developed a partial response (PR) after one year of vaccine treatment, whereas long-lasting stable disease (SD) ≥ 8.5 months was demonstrated in another six patients. The median overall survival (OS) was 25.9 months. Patients demonstrated significant improved OS (P = 0.03) when compared with the group of patients excluded because of HLA-A2 negativity. IDO-specific CD8(+) T-cell immunity was demonstrated by IFN-γ Elispot and Tetramer staining. Fluorescence-activated cell sorting analyses demonstrated a significant reduction of the Treg population (P = 0.03) after the sixth vaccine (2.5 months) compared with pretreatment levels. Furthermore, expression of IDO was detected in nine of ten tumor biopsies by immunohistochemistry. High-performance liquid chromatography analyses of kynurenine/tryptophan (Kyn/Trp) ratio in sera were performed. In long-term analyses of two clinical responding patients, the ratio of Kyn/Trp remained stable. CONCLUSIONS The vaccine was well tolerated with no severe toxicity occurring. A median OS of 25.9 months was demonstrated and long-lasting PR+SD was seen in 47% of the patients.
Collapse
MESH Headings
- Adenocarcinoma/enzymology
- Adenocarcinoma/mortality
- Adenocarcinoma/secondary
- Adenocarcinoma/therapy
- Adjuvants, Immunologic/administration & dosage
- Aged
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Carcinoma, Non-Small-Cell Lung/enzymology
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/secondary
- Carcinoma, Non-Small-Cell Lung/therapy
- Disease-Free Survival
- Female
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/blood
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Kaplan-Meier Estimate
- Kynurenine/blood
- Lung Neoplasms/enzymology
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Lymphatic Metastasis
- Male
- Middle Aged
- Statistics, Nonparametric
- T-Lymphocytes, Regulatory/enzymology
- T-Lymphocytes, Regulatory/immunology
- Treatment Outcome
- Tryptophan/blood
- Vaccination
Collapse
Affiliation(s)
- Trine Zeeberg Iversen
- Authors' Affiliations: Center for Cancer Immune Therapy, Department of Haematology & Department of Oncology, Copenhagen University Hospital at Herlev, Herlev; Department of Pathology, Bispebjerg Hospital, Team Bispebjerg (RH), Bispebjerg Bakke, Copenhagen NV, Denmark Interfaculty Institute of Biochemistry; and Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Larsen SK, Munir S, Woetmann A, Frøsig TM, Odum N, Svane IM, Becker JC, Andersen MH. Functional characterization of Foxp3-specific spontaneous immune responses. Leukemia 2013; 27:2332-40. [PMID: 23812418 DOI: 10.1038/leu.2013.196] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/20/2013] [Accepted: 06/24/2013] [Indexed: 12/25/2022]
Abstract
Tumor-infiltrating CD4+CD25+ regulatory T cells (Tregs) are associated with an impaired prognosis in several cancers. The transcription factor forkhead box P3 (Foxp3) is generally expressed in Tregs. Here, we identify and characterize spontaneous cytotoxic immune responses to Foxp3-expressing cells in peripheral blood of healthy volunteers and cancer patients. These immune responses were directed against a HLA-A2-restricted peptide epitope derived from Foxp3. Foxp3-reactive T cells were characterized as cytotoxic CD8+ T cells. These cells recognized dendritic cells incubated with recombinant Foxp3 protein indicating that this protein was indeed internalized, processed and cross-presented in the context of HLA-A2. More importantly, however, Foxp3-specific T cells were able to specifically recognize Tregs. Similarly, Foxp3+ malignant T cells established from a Cutaneous T-cell lymphomas (CTCL) patient were readily killed by the Foxp3-specific cytotoxic T lymphocytes. The spontaneous presence of Foxp3-specific cytotoxic T-cell responses suggest a general role of such T cells in the complex network of immune regulation as such responses may eliminate Tregs, that is, suppression of the suppressors. Consequently, induction of Foxp3-specific cytotoxic T-cell responses appears as an attractive tool to boost spontaneous or therapeutically provoked immune responses, for example, for the therapy of cancer.
Collapse
Affiliation(s)
- S K Larsen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, 54P4, Copenhagen University Hospital, Herlev, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kamperschroer C, O'Donnell LM, Schneider PA, Li D, Roy M, Coskran TM, Kawabata TT. Measuring T-cell responses against LCV and CMV in cynomolgus macaques using ELISPOT: potential application to non-clinical testing of immunomodulatory therapeutics. J Immunotoxicol 2013; 11:35-43. [PMID: 23461640 DOI: 10.3109/1547691x.2013.766287] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
A number of immunomodulatory therapeutics increase the risk of disease associated with latent herpesviruses such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV), a member of the lymphocryptovirus (LCV) family that infects humans. The diseases associated with loss of immunity to these viruses can have major impacts on patients as well as on the commercial viability of the immunomodulatory therapeutics. In an effort to develop non-clinical methods for measuring effects on anti-viral immunity, we have developed an interferon (IFN)-γ enzyme-linked immunosorbent spot (ELISPOT) assay to quantify the number of CMV or LCV-reactive T-cells in peripheral blood of cynomolgus macaques. After optimization of various parameters, the IFN-γ ELISPOT assay was characterized for specificity, intra-assay, monkey-to-monkey, and longitudinal variability and sensitivity to immunosuppression. The results show that nearly all animals have detectable responses against both CMV and LCV and responses were derived from T-cells specific to the virus of interest. Analyses of variability show assay reproducibility (≤23% CV), and that variability over time in anti-viral responses in individual animals (larger for LCV than for CMV) was ∼2-fold in most animals over a 3-month time period, which is predicted to allow for detection of drug-induced changes when using group sizes typical of non-clinical studies. In addition, the IFN-γ ELISPOT assay was capable of detecting decreases in the numbers of CMV and LCV reactive T-cells induced by immunosuppressive drugs in vitro. This assay may allow for non-clinical assessment of the effects of immunomodulatory therapeutics on anti-viral T-cell immunity in monkeys, and may help determine if therapeutics increase the risk of reactivating latent viral infections.
Collapse
Affiliation(s)
- Cris Kamperschroer
- Drug Safety Research and Development, Pfizer Global Research and Development , Pfizer, Inc., Groton, CT , USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Munir S, Andersen GH, Met Ö, Donia M, Frøsig TM, Larsen SK, Klausen TW, Svane IM, Andersen MH. HLA-restricted CTL that are specific for the immune checkpoint ligand PD-L1 occur with high frequency in cancer patients. Cancer Res 2013; 73:1764-76. [PMID: 23328583 DOI: 10.1158/0008-5472.can-12-3507] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PD-L1 (CD274) contributes to functional exhaustion of T cells and limits immune responses in patients with cancer. In this study, we report the identification of an human leukocyte antigen (HLA)-A2-restricted epitope from PD-L1, and we describe natural, cytolytic T-cell reactivity against PD-L1 in the peripheral blood of patients with cancer and healthy individuals. Notably, PD-L1-specific T cells were able not only to recognize and kill tumor cells but also PD-L1-expressing dendritic cells in a PD-L1-dependent manner, insofar as PD-L1 ablation rescued dendritic cells from killing. Furthermore, by incubating nonprofessional antigen-presenting cells with long peptides from PD-L1, we found that PD-L1 was rapidly internalized, processed, and cross-presented by HLA-A2 on the cell surface. Apparently, this cross-presentation was TAP-independent, as it was conducted not only by B cells but in addition by TAP-deficient T2-cells. This is intriguing, as soluble PD-L1 has been detected in the sera from patients with cancer. PD-L1-specific CTL may boost immunity by the killing of immunosuppressive tumor cells as well as regulatory cells. However, PD-L1-specific CTLs may as well suppress immunity by the elimination of normal immune cells especially PD-L1 expressing mature dendritic cells.
Collapse
Affiliation(s)
- Shamaila Munir
- Center for Cancer Immune Therapy, Department of Hematology and Oncology, Copenhagen University Hospital, Herlev, Herlev Ringvej, Herlev, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ellebaek E, Engell-Noerregaard L, Iversen TZ, Froesig TM, Munir S, Hadrup SR, Andersen MH, Svane IM. Metastatic melanoma patients treated with dendritic cell vaccination, Interleukin-2 and metronomic cyclophosphamide: results from a phase II trial. Cancer Immunol Immunother 2012; 61:1791-804. [PMID: 22426890 PMCID: PMC11029126 DOI: 10.1007/s00262-012-1242-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 03/04/2012] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DC) are the most potent antigen presenting cells and have proven effective in stimulation of specific immune responses in vivo. Competing immune inhibition could limit the clinical efficacy of DC vaccination. In this phase II trial, metronomic Cyclophosphamide and a Cox-2 inhibitor have been added to a DC vaccine with the intend to dampen immunosuppressive mechanisms. Twenty-eight patients with progressive metastatic melanoma were treated with autologous DCs pulsed with survivin, hTERT, and p53-derived peptides (HLA-A2(+)) or tumor lysate (HLA-A2(-)). Concomitantly the patients were treated with IL-2, Cyclophosphamide, and Celecoxib. The treatment was safe and tolerable. Sixteen patients (57 %) achieved stable disease (SD) at 1st evaluation and 8 patients had prolonged SD (7-13.7 months). The median OS was 9.4 months. Patients with SD had an OS of 10.5 months while patients with progressive disease (PD) had an OS of 6.0 months (p = 0.048) even though there were no differences in prognostic factors between the two groups. Despite the use of metronomic Cyclophosphamide, regulatory T cells did not decrease during treatment. Indirect IFN-γ ELISPOT assays showed a general increase in immune responses from baseline to the time of 4th vaccination. Induction of antigen-specific immune responses was seen in 9 out of 15 screened HLA-A2(+) patients. In conclusion, the number of patients obtaining SD more than doubled and 6-month survival significantly increased compared to a previous trial without Cyclophosphamide and Celecoxib. A general increase in immune responses against the tested peptides was observed.
Collapse
Affiliation(s)
- Eva Ellebaek
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Lotte Engell-Noerregaard
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Trine Zeeberg Iversen
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Thomas Moerch Froesig
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Pharmacology and Pharmacotherapy, Pharmaceutical Faculty, University of Copenhagen, Copenhagen, Denmark
| | - Shamaila Munir
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Sine Reker Hadrup
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Mads Hald Andersen
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Inge Marie Svane
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| |
Collapse
|
34
|
Use of ESAT-6-CFP-10 fusion protein in the bovine interferon-gamma ELISPOT assay for diagnosis of Mycobacterium bovis infection in cattle. J Microbiol Methods 2012; 90:298-304. [DOI: 10.1016/j.mimet.2012.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/25/2012] [Accepted: 06/01/2012] [Indexed: 12/17/2022]
|
35
|
Yang T, Chen ZZ, Kolb HJ, Buhmann R. A novel nonradioactive CFDA assay to monitor the cellular immune response in myeloid leukemia. Immunobiology 2012; 218:548-53. [PMID: 22883564 DOI: 10.1016/j.imbio.2012.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 06/24/2012] [Accepted: 06/26/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND Donor lymphocyte transfusion (DLT) may induce the graft-versus-leukemia (GVL) effect for patients with AML relapsed after transplant. However, AML is a highly diverse disease and the limited overall efficacy of DLT in clinical practice emphasizes the importance of identifying a specific subgroup of patients who might benefit from this treatment approach. OBJECTIVE To monitor the cellular immune response after DLT, we developed an active specific immunization strategy using in vitro generated AML-trained T cells to induce a highly specific antileukemic T-cell response and thus established a novel nonradioactive assay system to assess the antileukemia immunity by flow cytometry, correlated with [3H]-thymidine uptake. METHODS The myeloid blasts derived from five patients with AML relapsed post-allogeneic hematopoietic stem cell transplantation (allo-HSCT) were first labeled with CFDA (5,6-carboxyfluorescein diacetate succinimidyl ester). To analyze the growth inhibitory potential of the donor T cells trained by AML progenitor cells, the myeloid blasts were induced to proliferate by means of a cytokine cocktail (50ng/mL of SCF; 25ng/mL of IL-3; 100ng/mL of GM-CSF; 100ng/mL of G-CSF; 2U/mL of EPO; 0.47g/L of transferrin; and 5×10(-5)mmol/L of 2-ME). The T cell mediated growth inhibitory potential was detected after 5 days by flow cytometry and correlated with [3H]-thymidine uptake. The simultaneous use of TO-PRO-dye and calibrate beads allowed not only the cell viability to be known but also allowed quantification of the effector function. RESULTS Here, we applied a CFDA dye to track the proliferation and expansion of AML blasts in response to the cytokine cocktail in vitro. AML-trained T cells, expressed high levels of the activation markers CD25 and CD69, and were generated to recognize the leukemic progenitor cells and inhibit cytokine-induced leukemic cell proliferation, which is an active specific immunization strategy circumventing the identification of leukemia-associated antigens. The capability of proliferation inhibition of AML-trained T cells evaluated with our nonradioactive, CFDA-based assay provided comparable results with the classic [3H]-thymidine assay with an even lower ratio of effector to target cells. CONCLUSION Taken together, the novel, nonradioactive, CFDA-based assay was a robust tool to monitor the antileukemic immune response after DLT in myeloid leukemias.
Collapse
Affiliation(s)
- Ting Yang
- Department of Hematology, Union Hospital, Fujian Medical University, Fuzhou, China
| | | | | | | |
Collapse
|
36
|
Characterization of Spontaneous Immune Responses against Long Peptides Derived from Bcl-X(L) in Cancer Patients Using Elispot. Cells 2012; 1:51-60. [PMID: 24710413 PMCID: PMC3901095 DOI: 10.3390/cells1020051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Revised: 03/07/2012] [Accepted: 04/03/2012] [Indexed: 11/23/2022] Open
Abstract
In recent years we and others have used the ELISPOT assay successfully to identify novel tumor antigens by the characterization of spontaneous HLA class I restricted immune responses against a number of minimal 9–10 amino acid long peptide epitopes. In the present study, we examined the capability of using longer peptides when scrutinizing Peripheral Blood Mononuclear Cells (PMBC) from melanoma patients for spontaneous immunity by means of ELISPOT IFN-γ secretion assay. To this end, we examined PBMC for the presence of specific T-cell responses against long peptides derived from the tumor associated antigen BCL-X(L). The protein product of the larger BCL-X(L) differs from Bcl-X(S) protein by an inserted region (amino acids 126–188). Thus, we scrutinized eight long peptides covering this inserted region for spontaneous immunity. The peptides were overlapping and consisted of 20–23 amino acids. PBMC were pre-stimulated with peptide-pulsed autologous dendritic cells (DC) and subjected to the IFN-γ ELISPOT assay. Four of the BCL-X(L) derived peptides elicited very frequent responses in several patients. Additionally, in all patients responses against more than one of the peptides could be detected. In conclusion several long BCL-X(L) derived peptide epitopes exist, which may be used in anti-cancer immunity. Furthermore, the ELISPOT assay offers an attractive and sensitive method for the characterization of spontaneous immune reactivity against long peptides.
Collapse
|
37
|
Munir S, Larsen SK, Iversen TZ, Donia M, Klausen TW, Svane IM, Straten PT, Andersen MH. Natural CD4+ T-cell responses against indoleamine 2,3-dioxygenase. PLoS One 2012; 7:e34568. [PMID: 22539948 PMCID: PMC3335144 DOI: 10.1371/journal.pone.0034568] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 03/02/2012] [Indexed: 01/14/2023] Open
Abstract
Background The enzyme indoleamine 2,3-dioxygenase (IDO) contributes to immune tolerance in a variety of settings. In cancer IDO is expressed within the tumor itself as well as in antigen-presenting cells in tumor-draining lymph nodes, where it endorses the establishment of peripheral immune tolerance to tumor antigens. Recently, we described cytotoxic CD8+ T-cell reactivity towards IDO-derived peptides. Methods and Findings In the present study, we show that CD4+ helper T cells additionally spontaneously recognize IDO. Hence, we scrutinized the vicinity of the previously described HLA-A*0201-restricted IDO-epitope for CD4+ T-cell epitopes. We demonstrated the presence of naturally occurring IDO-specific CD4+ T cells in cancer patients and to a lesser extent in healthy donors by cytokine release ELISPOT. IDO-reactive CD4+ T cells released IFN-γ, TNF-α, as well as IL-17. We confirm HLA class II-restriction by the addition of HLA class II specific blocking antibodies. In addition, we detected a trend between class I- and class II-restricted IDO responses and detected an association between IDO-specific CD4+ T cells and CD8+ CMV-responses. Finally, we could detect IL-10 releasing IDO-reactive CD4+ T cells. Conclusion IDO is spontaneously recognized by HLA class II-restricted, CD4+ T cells in cancer patients and in healthy individuals. IDO-specific T cells may participate in immune-regulatory networks where the activation of pro-inflammatory IDO-specific CD4+ responses may well overcome or delay the immune suppressive actions of the IDO-protein, which are otherwise a consequence of the early expression of IDO in maturing antigen presenting cells. In contrast, IDO-specific regulatory T cells may enhance IDO-mediated immune suppression.
Collapse
Affiliation(s)
- Shamaila Munir
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Stine Kiaer Larsen
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Trine Zeeberg Iversen
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Marco Donia
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
- Department of Biomedical Sciences, University of Catania, Catania, Italy
| | - Tobias Wirenfeldt Klausen
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Inge Marie Svane
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Per thor Straten
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - Mads Hald Andersen
- Department of Hematology and Oncology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Herlev, Denmark
- * E-mail:
| |
Collapse
|
38
|
Kamperschroer C, Kaur A, Lebrec H. A summary of meeting proceedings for ‘Measuring immune responses in non-human primates for drug development—Opportunities and challenges for predicting human efficacy and immunotoxicity’. J Immunotoxicol 2012; 9:108-20. [DOI: 10.3109/1547691x.2011.631610] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
39
|
Gupta NK, Tomar P, Sharma V, Dixit VK. Development and characterization of chitosan coated poly-(ɛ-caprolactone) nanoparticulate system for effective immunization against influenza. Vaccine 2011; 29:9026-37. [DOI: 10.1016/j.vaccine.2011.09.033] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/31/2011] [Accepted: 09/09/2011] [Indexed: 10/17/2022]
|
40
|
Raddassi K, Kent SC, Yang J, Bourcier K, Bradshaw EM, Seyfert-Margolis V, Nepom GT, Kwok WW, Hafler DA. Increased frequencies of myelin oligodendrocyte glycoprotein/MHC class II-binding CD4 cells in patients with multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2011; 187:1039-46. [PMID: 21653833 DOI: 10.4049/jimmunol.1001543] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by infiltration of pathogenic immune cells in the CNS resulting in destruction of the myelin sheath and surrounding axons. We and others have previously measured the frequency of human myelin-reactive T cells in peripheral blood. Using T cell cloning techniques, a modest increase in the frequency of myelin-reactive T cells in patients as compared with control subjects was observed. In this study, we investigated whether myelin oligodendrocyte glycoprotein (MOG)-specific T cells could be detected and their frequency was measured using DRB1*0401/MOG(97-109(107E-S)) tetramers in MS subjects and healthy controls expressing HLA class II DRB1*0401. We defined the optimal culture conditions for expansion of MOG-reactive T cells upon MOG peptide stimulation of PMBCs. MOG(97-109)-reactive CD4(+) T cells, isolated with DRB1*0401/MOG(97-109) tetramers, and after a short-term culture of PMBCs with MOG(97-109) peptides, were detected more frequently from patients with MS as compared with healthy controls. T cell clones from single cell cloning of DRB1*0401/MOG(97-109(107E-S)) tetramer(+) cells confirmed that these T cell clones were responsive to both the native and the substituted MOG peptide. These data indicate that autoantigen-specific T cells can be detected and enumerated from the blood of subjects using class II tetramers, and the frequency of MOG(97-109)-reactive T cells is greater in patients with MS as compared with healthy controls.
Collapse
Affiliation(s)
- Khadir Raddassi
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510-8018, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Indoleamine 2,3-dioxygenase (IDO) is an immunoregulatory enzyme that is implicated in suppressing T-cell immunity in normal and pathologic settings. Here, we describe that spontaneous cytotoxic T-cell reactivity against IDO exists not only in patients with cancer but also in healthy persons. We show that the presence of such IDO-specific CD8(+) T cells boosted T-cell immunity against viral or tumor-associated antigens by eliminating IDO(+) suppressive cells. This had profound effects on the balance between interleukin-17 (IL-17)-producing CD4(+) T cells and regulatory T cells. Furthermore, this caused an increase in the production of the proinflammatory cytokines IL-6 and tumor necrosis factor-α while decreasing the IL-10 production. Finally, the addition of IDO-inducing agents (ie, the TLR9 ligand cytosine-phosphate-guanosine, soluble cytotoxic T lymphocyte-associated antigen 4, or interferon γ) induced IDO-specific T cells among peripheral blood mononuclear cells from patients with cancer as well as healthy donors. In the clinical setting, IDO may serve as an important and widely applicable target for immunotherapeutic strategies in which IDO plays a significant regulatory role. We describe for the first time effector T cells with a general regulatory function that may play a vital role for the mounting or maintaining of an effective adaptive immune response. We suggest terming such effector T cells "supporter T cells."
Collapse
|
42
|
Identification of a cyclin B1-derived CTL epitope eliciting spontaneous responses in both cancer patients and healthy donors. Cancer Immunol Immunother 2010; 60:227-34. [PMID: 20981424 PMCID: PMC3024510 DOI: 10.1007/s00262-010-0933-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 09/18/2010] [Indexed: 11/03/2022]
Abstract
With the aim to identify cyclin B1-derived peptides with high affinity for HLA-A2, we used three in silico prediction algorithms to screen the protein sequence for possible HLA-A2 binders. One peptide scored highest in all three algorithms, and the high HLA-A2-binding affinity of this peptide was verified in an HLA stabilization assay. By stimulation with peptide-loaded dendritic cells a CTL clone was established, which was able to kill two breast cancer cell lines in an HLA-A2-dependent and peptide-specific manner, demonstrating presentation of the peptide on the surface of cancer cells. Furthermore, blood from cancer patients and healthy donors was screened for spontaneous T-cell reactivity against the peptide in IFN-γ ELISPOT assays. Patients with breast cancer, malignant melanoma, or renal cell carcinoma hosted powerful and high-frequency T-cell responses against the peptide. In addition, when blood from healthy donors was tested, similar responses were observed. Ultimately, serum from cancer patients and healthy donors was analyzed for anti-cyclin B1 antibodies. Humoral responses against cyclin B1 were frequently detected in both cancer patients and healthy donors. In conclusion, a high-affinity cyclin B1-derived HLA-A2-restricted CTL epitope was identified, which was presented on the cell surface of cancer cells, and elicited spontaneous T-cell responses in cancer patients and healthy donors.
Collapse
|
43
|
Moodie Z, Price L, Gouttefangeas C, Mander A, Janetzki S, Löwer M, Welters MJP, Ottensmeier C, van der Burg SH, Britten CM. Response definition criteria for ELISPOT assays revisited. Cancer Immunol Immunother 2010; 59:1489-501. [PMID: 20549207 PMCID: PMC2909425 DOI: 10.1007/s00262-010-0875-4] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 05/31/2010] [Indexed: 11/28/2022]
Abstract
No consensus has been reached on how to determine if an immune response has been detected based on raw data from an ELISPOT assay. The goal of this paper is to enable investigators to understand and readily implement currently available methods for response determination. We describe empirical and statistical approaches, identifying the strengths and limitations of each approach to allow readers to rationally select and apply a scientifically sound method appropriate to their specific laboratory setting. Five representative approaches were applied to data sets from the CIMT Immunoguiding Program and the response detection and false positive rates were compared. Simulation studies were also performed to compare empirical and statistical approaches. Based on these, we recommend the use of a non-parametric statistical test. Further, we recommend that six medium control wells or four wells each for both medium control and experimental conditions be performed to increase the sensitivity in detecting a response, that replicates with large variation in spot counts be filtered out, and that positive responses arising from experimental spot counts below the estimated limit of detection be interpreted with caution. Moreover, a web-based user interface was developed to allow easy access to the recommended statistical methods. This interface allows the user to upload data from an ELISPOT assay and obtain an output file of the binary responses.
Collapse
Affiliation(s)
- Z Moodie
- Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
High immunogenic potential of p53 mRNA-transfected dendritic cells in patients with primary breast cancer. Breast Cancer Res Treat 2010; 125:395-406. [DOI: 10.1007/s10549-010-0844-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 03/10/2010] [Indexed: 01/14/2023]
|
45
|
Lv Y, Ruan Z, Wang L, Ni B, Wu Y. Identification of a novel conserved HLA-A*0201-restricted epitope from the spike protein of SARS-CoV. BMC Immunol 2009; 10:61. [PMID: 19958537 PMCID: PMC2792222 DOI: 10.1186/1471-2172-10-61] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 12/03/2009] [Indexed: 01/10/2023] Open
Abstract
Background The spike (S) protein is a major structural glycoprotein of coronavirus (CoV), the causal agent of severe acute respiratory syndrome (SARS). The S protein is a potent target for SARS-specific cell-mediated immune responses. However, the mechanism CoV pathogenesis in SARS and the role of special CTLs in virus clearance are still largely uncharacterized. Here, we describe a study that leads to the identification of a novel HLA-A*0201-restricted epitope from conserved regions of S protein. Results First, different SARS-CoV sequences were analyzed to predict eight candidate peptides from conserved regions of the S protein based upon HLA-A*0201 binding and proteosomal cleavage. Four of eight candidate peptides were tested by HLA-A*0201 binding assays. Among the four candidate peptides, Sp8 (S958-966, VLNDILSRL) induced specific CTLs both ex vivo in PBLs of healthy HLA-A2+ donors and in HLA-A2.1/Kb transgenic mice immunized with a plasmid encoding full-length S protein. The immunized mice released IFN-γ and lysed target cells upon stimulation with Sp8 peptide-pulsed autologous dendritic cells in comparison to other candidates. Conclusion These results suggest that Sp8 is a naturally processed epitope. We propose that Sp8 epitope should help in the characterization of mechanisms of virus control and immunopathology in SARS-CoV infection.
Collapse
Affiliation(s)
- Yanbo Lv
- Institute of Immunology, PLA, Third Military Medical University, 30 Gaotanyan Street, Chongqing 400038, PR China.
| | | | | | | | | |
Collapse
|
46
|
Andersen RS, Srensen RB, Andersen MH, Straten PT. CD8 T-Cell Responses Against Cyclin B1. Clin Cancer Res 2009. [DOI: 10.1158/1078-0432.ccr-09-2097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Rikke Sick Andersen
- Center for Cancer Immune Therapy, Department of Hematology, University Hospital Herlev, Herlev, Denmark
| | - Rikke Bk Srensen
- Center for Cancer Immune Therapy, Department of Hematology, University Hospital Herlev, Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy, Department of Hematology, University Hospital Herlev, Herlev, Denmark
| | - Per thor Straten
- Center for Cancer Immune Therapy, Department of Hematology, University Hospital Herlev, Herlev, Denmark
| |
Collapse
|
47
|
Køllgaard T, Duval L, Schmidt H, Kaltoft K, Seremet T, Andersen MH, Maase HVD, Straten PT, Hadrup SR. Longitudinal immune monitoring of patients receiving intratumoral injection of a MART-1 T-cell receptor-transduced cell line (C-Cure 709). Cytotherapy 2009; 11:631-41. [PMID: 19530030 DOI: 10.1080/14653240902923146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND AIMS Adoptive transfer of tumor-specific lymphocytes is a promising strategy in the treatment of cancer. We conducted intratumoral administration of an allogeneic irradiated continuous T-cell line (C-Cure 709) expressing an HLA-A2-restricted MART-1-specific T-cell receptor (TCR) into HLA-A2(+) melanoma patients. The C-Cure 709 cell line is cytotoxic against MART-1(+) HLA-A2(+) melanoma cell lines and secretes several immune stimulatory cytokines upon stimulation. METHODS Anti-tumor immune responses against the commonly expressed tumor antigen (Ag) MART-1 were longitudinally analyzed in peripheral blood by fluorescence-activated cell sorting (FACS) before and after intratumoral injection of C-Cure 709. RESULTS No treatment-induced increase in Ag-specific T-cell frequencies was observed in peripheral blood, and the phenotype of MART-1-specific T cells was very stable during the treatment. Interestingly, despite a very stable frequency of MART-1-specific T cells over the course of treatment, clonotype mapping revealed that the response was in fact highly diverse and dynamic, with new clonotypes emerging during treatment. Only a few clonotypes were recurrently detected in consecutive samples. One MART-1-specific T-cell clone disappearing from peripheral blood was later detected in a metastatic lesion. CONCLUSIONS Sequence analyzes of the CDR3 region revealed conserved structural characteristics in the MART-1-specific TCR used by T-cell clones.
Collapse
Affiliation(s)
- Tania Køllgaard
- Center for Cancer Immune Therapy (CCIT), University Hospital Herlev, Herlev, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
The immune system strikes back: cellular immune responses against indoleamine 2,3-dioxygenase. PLoS One 2009; 4:e6910. [PMID: 19738905 PMCID: PMC2734165 DOI: 10.1371/journal.pone.0006910] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 06/26/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The enzyme indoleamine 2,3-dioxygenase (IDO) exerts an well established immunosuppressive function in cancer. IDO is expressed within the tumor itself as well as in antigen-presenting cells in tumor-draining lymph nodes, where it promotes the establishment of peripheral immune tolerance to tumor antigens. In the present study, we tested the notion whether IDO itself may be subject to immune responses. METHODS AND FINDINGS The presence of naturally occurring IDO-specific CD8 T cells in cancer patients was determined by MHC/peptide stainings as well as ELISPOT. Antigen specific cytotoxic T lymphocytes (CTL) from the peripheral blood of cancer patients were cloned and expanded. The functional capacity of the established CTL clones was examined by chrome release assays. The study unveiled spontaneous cytotoxic T-cell reactivity against IDO in peripheral blood as well as in the tumor microenvironment of different cancer patients. We demonstrate that these IDO reactive T cells are indeed peptide specific, cytotoxic effector cells. Hence, IDO reactive T cells are able to recognize and kill tumor cells including directly isolated AML blasts as well as IDO-expressing dendritic cells, i.e. one of the major immune suppressive cell populations. CONCLUSION IDO may serve as an important and widely applicable target for anti-cancer immunotherapeutic strategies. Furthermore, as emerging evidence suggests that IDO constitutes a significant counter-regulatory mechanism induced by pro-inflammatory signals, IDO-based immunotherapy holds the promise to boost anti-cancer immunotherapy in general.
Collapse
|
49
|
Andersen MH, Sørensen RB, Brimnes MK, Svane IM, Becker JC, thor Straten P. Identification of heme oxygenase-1-specific regulatory CD8+ T cells in cancer patients. J Clin Invest 2009; 119:2245-56. [PMID: 19662679 DOI: 10.1172/jci38739] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Treg deficiencies are associated with autoimmunity. Conversely, CD4+ and CD8+ Tregs accumulate in the tumor microenvironment and are associated with prevention of antitumor immunity and anticancer immunotherapy. Recently, CD4+ Tregs have been much studied, but little is known about CD8+ Tregs and the antigens they recognize. Here, we describe what we believe to be the first natural target for CD8+ Tregs. Naturally occurring HLA-A2-restricted CD8+ T cells specific for the antiinflammatory molecule heme oxygenase-1 (HO-1) were able to suppress cellular immune responses with outstanding efficacy. HO-1-specific CD8+ T cells were detected ex vivo and in situ among T cells from cancer patients. HO-1-specific T cells isolated from the peripheral blood of cancer patients inhibited cytokine release, proliferation, and cytotoxicity of other immune cells. Notably, the inhibitory effect of HO-1-specific T cells was far more pronounced than that of conventional CD4+CD25+CD127- Tregs. The inhibitory activity of HO-1-specific T cells seemed at least partly to be mediated by soluble factors. Our data link the cellular stress response to the regulation of adaptive immunity, expand the role of HO-1 in T cell-mediated immunoregulation, and establish a role for peptide-specific CD8+ T cells in regulating cellular immune responses. Identification of potent antigen-specific CD8+ Tregs may open new avenues for therapeutic interventions in both autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Mads Hald Andersen
- Center for Cancer Immune Therapy, Department of Hematology, Herlev University Hospital, Herlev, Denmark.
| | | | | | | | | | | |
Collapse
|
50
|
Givan AL. A Flow Cytometric Assay for Quantitation of Rare Antigen-Specific T Cells: Using Cell-Tracking Dyes to Calculate Precursor Frequencies for Proliferation. Immunol Invest 2009; 36:563-80. [DOI: 10.1080/08820130701683803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|