1
|
Gille I, Hagedoorn RS, van der Meer-Prins EMW, Heemskerk MHM, Heidt S. Chimeric HLA antibody receptor T cells to target HLA-specific B cells in solid organ transplantation. HLA 2023; 102:436-448. [PMID: 37370222 DOI: 10.1111/tan.15146] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/19/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023]
Abstract
HLA-sensitized patients on the transplant waiting list harbor antibodies and memory B cells directed against allogeneic HLA molecules, which decreases the chance to receive a compatible donor organ. Current desensitization strategies non-specifically target circulating antibodies and B cells, warranting the development of therapies that specifically affect HLA-directed humoral immune responses. We developed Chimeric HLA Antibody Receptor (CHAR) constructs comprising the extracellular part of HLA-A2 or HLA-A3 coupled to CD28-CD3ζ domains. CHAR-transduced cells expressing reporter constructs encoding T-cell activation markers, and CHAR-transduced CD8+ T cells from healthy donors were stimulated with HLA-specific monoclonal antibody-coated microbeads, and HLA-specific B cell hybridomas. CHAR T cell activation was measured by upregulation of T cell activation markers and IFNγ secretion, whereas CHAR T cell killing of B cell hybridomas was assessed in chromium release assays and by IgG ELISpot. HLA-A2- and HLA-A3-CHAR expressing cells were specifically activated by HLA-A2- and HLA-A3-specific monoclonal antibodies, either soluble or coated on microbeads, as shown by CHAR-induced transcription factors. HLA-A2 and HLA-A3 CHAR T cells efficiently produced IFNγ with exquisite specificity and were capable of specifically lysing hybridoma cells expressing HLA-A2- or HLA-A3-specific B-cell receptors, respectively. Finally, we mutated the α3 domain of the CHAR molecules to minimize any alloreactive T-cell reactivity against CHAR T cells, while retaining CHAR activity. These data show proof of principle for CHAR T cells to serve as precision immunotherapy to specifically desensitize (highly) sensitized solid organ transplant candidates and to treat antibody-mediated rejection after solid organ transplantation.
Collapse
Affiliation(s)
- Ilse Gille
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Renate S Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
2
|
Kandasamy M, Gileadi U, Rijal P, Tan TK, Lee LN, Chen J, Prota G, Klenerman P, Townsend A, Cerundolo V. Recombinant single-cycle influenza virus with exchangeable pseudotypes allows repeated immunization to augment anti-tumour immunity with immune checkpoint inhibitors. eLife 2023; 12:76414. [PMID: 36626205 PMCID: PMC9831609 DOI: 10.7554/elife.76414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/08/2022] [Indexed: 01/11/2023] Open
Abstract
Virus-based tumour vaccines offer many advantages compared to other antigen-delivering systems. They generate concerted innate and adaptive immune response, and robust CD8+ T cell responses. We engineered a non-replicating pseudotyped influenza virus (S-FLU) to deliver the well-known cancer testis antigen, NY-ESO-1 (NY-ESO-1 S-FLU). Intranasal or intramuscular immunization of NY-ESO-1 S-FLU virus in mice elicited a strong NY-ESO-1-specific CD8+ T cell response in lungs and spleen that resulted in the regression of NY-ESO-1-expressing lung tumour and subcutaneous tumour, respectively. Combined administration with anti-PD-1 antibody, NY-ESO-1 S-FLU virus augmented the tumour protection by reducing the tumour metastasis. We propose that the antigen delivery through S-FLU is highly efficient in inducing antigen-specific CD8+ T cell response and protection against tumour development in combination with PD-1 blockade.
Collapse
Affiliation(s)
- Matheswaran Kandasamy
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Pramila Rijal
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Tiong Kit Tan
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Lian N Lee
- Nuffield Department of Medicine and Translational Gastroenterology Unit, Peter Medawar Building, University of OxfordOxfordUnited Kingdom
| | - Jili Chen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Gennaro Prota
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine and Translational Gastroenterology Unit, Peter Medawar Building, University of OxfordOxfordUnited Kingdom
| | - Alain Townsend
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
3
|
Immisch L, Papafotiou G, Popp O, Mertins P, Blankenstein T, Willimsky G. H3.3K27M mutation is not a suitable target for immunotherapy in HLA-A2 + patients with diffuse midline glioma. J Immunother Cancer 2022; 10:jitc-2022-005535. [PMID: 36302563 PMCID: PMC9621174 DOI: 10.1136/jitc-2022-005535] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Diffuse midline glioma is the leading cause of solid cancer-related deaths in children with very limited treatment options. A majority of the tumors carry a point mutation in the histone 3 variant (H3.3) creating a potential HLA-A*02:01 binding epitope (H3.3K27M26-35). Here, we isolated an H3.3K27M-specific T cell receptor (TCR) from transgenic mice expressing a diverse human TCR repertoire. Despite a high functional avidity of H3.3K27M-specific T cells, we were not able to achieve recognition of cells naturally expressing the H3.3K27M mutation, even when overexpressed as a transgene. Similar results were obtained with T cells expressing the published TCR 1H5 against the same epitope. CRISPR/Cas9 editing was used to exclude interference by endogenous TCRs in donor T cells. Overall, our data provide strong evidence that the H3.3K27M mutation is not a suitable target for cancer immunotherapy, most likely due to insufficient epitope processing and/or amount to be recognized by HLA-A*02:01 restricted CD8+ T cells.
Collapse
Affiliation(s)
- Lena Immisch
- Institute of Immunology, Charité Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,German Cancer Research Center, Heidelberg, Germany,German Cancer Consortium, partner site Berlin, Berlin, Germany
| | - George Papafotiou
- Institute of Immunology, Charité Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,German Cancer Research Center, Heidelberg, Germany,German Cancer Consortium, partner site Berlin, Berlin, Germany
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany,Berlin Institute of Health (BIH), Berlin, Germany
| | - Thomas Blankenstein
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany,Berlin Institute of Health (BIH), Berlin, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,German Cancer Research Center, Heidelberg, Germany,German Cancer Consortium, partner site Berlin, Berlin, Germany
| |
Collapse
|
4
|
Souter MN, Awad W, Li S, Pediongco TJ, Meehan BS, Meehan LJ, Tian Z, Zhao Z, Wang H, Nelson A, Le Nours J, Khandokar Y, Praveena T, Wubben J, Lin J, Sullivan LC, Lovrecz GO, Mak JY, Liu L, Kostenko L, Kedzierska K, Corbett AJ, Fairlie DP, Brooks AG, Gherardin NA, Uldrich AP, Chen Z, Rossjohn J, Godfrey DI, McCluskey J, Pellicci DG, Eckle SB. CD8 coreceptor engagement of MR1 enhances antigen responsiveness by human MAIT and other MR1-reactive T cells. J Exp Med 2022; 219:213423. [PMID: 36018322 PMCID: PMC9424912 DOI: 10.1084/jem.20210828] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/24/2022] [Accepted: 07/21/2022] [Indexed: 11/04/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells detect microbial infection via recognition of riboflavin-based antigens presented by the major histocompatibility complex class I (MHC-I)-related protein 1 (MR1). Most MAIT cells in human peripheral blood express CD8αα or CD8αβ coreceptors, and the binding site for CD8 on MHC-I molecules is relatively conserved in MR1. Yet, there is no direct evidence of CD8 interacting with MR1 or the functional consequences thereof. Similarly, the role of CD8αα in lymphocyte function remains ill-defined. Here, using newly developed MR1 tetramers, mutated at the CD8 binding site, and by determining the crystal structure of MR1-CD8αα, we show that CD8 engaged MR1, analogous to how it engages MHC-I molecules. CD8αα and CD8αβ enhanced MR1 binding and cytokine production by MAIT cells. Moreover, the CD8-MR1 interaction was critical for the recognition of folate-derived antigens by other MR1-reactive T cells. Together, our findings suggest that both CD8αα and CD8αβ act as functional coreceptors for MAIT and other MR1-reactive T cells.
Collapse
Affiliation(s)
- Michael N.T. Souter
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Wael Awad
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Shihan Li
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Troi J. Pediongco
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Bronwyn S. Meehan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Lucy J. Meehan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Zehua Tian
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Zhe Zhao
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Huimeng Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Adam Nelson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Yogesh Khandokar
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - T. Praveena
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Jacinta Wubben
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Jie Lin
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Lucy C. Sullivan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - George O. Lovrecz
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Melbourne, Australia
| | - Jeffrey Y.W. Mak
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ligong Liu
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Lyudmila Kostenko
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David P. Fairlie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Andrew G. Brooks
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Nicholas A. Gherardin
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Adam P. Uldrich
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Zhenjun Chen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia,Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Daniel G. Pellicci
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia,Murdoch Children’s Research Institute, Parkville, Melbourne, Australia
| | - Sidonia B.G. Eckle
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
5
|
Shenderov E, Kandasamy M, Gileadi U, Chen J, Shepherd D, Gibbs J, Prota G, Silk JD, Yewdell JW, Cerundolo V. Generation and characterization of HLA-A2 transgenic mice expressing the human TCR 1G4 specific for the HLA-A2 restricted NY-ESO-1 157-165 tumor-specific peptide. J Immunother Cancer 2021; 9:jitc-2021-002544. [PMID: 34088742 PMCID: PMC8183295 DOI: 10.1136/jitc-2021-002544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2021] [Indexed: 01/07/2023] Open
Abstract
Background NY-ESO-1 is a tumor-specific, highly immunogenic, human germ cell antigen of the MAGE-1 family that is a promising vaccine and cell therapy candidate in clinical trial development. The mouse genome does not encode an NY-ESO-1 homolog thereby not subjecting transgenic T-cells to thymic tolerance mechanisms that might impair in-vivo studies. We hypothesized that an NY-ESO-1 T cell receptor (TCR) transgenic mouse would provide the unique opportunity to study avidity of TCR response against NY-ESO-1 for tumor vaccine and cellular therapy development against this clinically relevant and physiological human antigen. Methods To study in vitro and in vivo the requirements for shaping an effective T cell response against the clinically relevant NY-ESO-1, we generated a C57BL/6 HLA-A*0201 background TCR transgenic mouse encoding the 1G4 TCR specific for the human HLA-A2 restricted, NY-ESO-1157-165 SLLMWITQC (9C), initially identified in an NY-ESO-1 positive melanoma patient. Results The HLA-A*0201 restricted TCR was positively selected on both CD4+ and CD8+ cells. Mouse 1G4 T cells were not activated by endogenous autoimmune targets or a large library of non-cognate viral antigens. In contrast, their activation by HLA-A2 NY-ESO-1157-165 complexes was evident by proliferation, CD69 upregulation, interferon-γ production, and interleukin-2 production, and could be tuned using a twofold higher affinity altered peptide ligand, NY-ESO-1157-165V. NY-ESO-1157-165V recombinant vaccination of syngeneic mice adoptively transferred with m1G4 CD8+ T cells controlled tumor growth in vivo. 1G4 transgenic mice suppressed growth of syngeneic methylcholanthrene (MCA) induced HHD tumor cells expressing the full-length human NY-ESO-1 protein but not MCA HHD tumor cells lacking NY-ESO-1. Conclusions The 1G4 TCR mouse model for the physiological human TCR against the clinically relevant antigen, NY-ESO-1, is a valuable tool with the potential to accelerate clinical development of NY-ESO-1-targeted T-cell and vaccine therapies.
Collapse
Affiliation(s)
- Eugene Shenderov
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK .,National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Matheswaran Kandasamy
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Jili Chen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Dawn Shepherd
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - James Gibbs
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gennaro Prota
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Jonathan D Silk
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK.,Next Generation Research, Adaptimmune, Abingdon, UK
| | - Jonathan W Yewdell
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| |
Collapse
|
6
|
Painter GF, Burn OK, Hermans IF. Using agonists for iNKT cells in cancer therapy. Mol Immunol 2020; 130:1-6. [PMID: 33340930 DOI: 10.1016/j.molimm.2020.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 01/03/2023]
Abstract
The capacity of α-galactosylceramide (α-GalCer) to act as an anti-cancer agent in mice through the specific stimulation of type I NKT (iNKT) cells has prompted extensive investigation to translate this finding to the clinic. However, low frequencies of iNKT cells in cancer patients and their hypo-responsiveness to repeated stimulation have been seen as barriers to its efficacy. Currently the most promising clinical application of α-GalCer, or its derivatives, is as stimuli for ex vivo expansion of iNKT cells for adoptive therapy, although some encouraging clinical results have recently been reported using α-GalCer pulsed onto large numbers of antigen presenting cells (APCs). In on-going preclinical studies, attempts to improve efficacy of injected iNKT cell agonists have focussed on optimising presentation in vivo, through encapsulation in particulate vectors, making structural changes that help binding to the presenting molecule CD1d, or injecting agonists covalently attached to recombinant CD1d. Variations on these same approaches are being used to enhance the APC-licencing function of iNKT cells in vivo to induce adaptive immune responses to associated tumour antigens. Looking ahead, a unique capacity of in vivo-activated iNKT cells to facilitate formation of resident memory CD8+ T cells is a new observation that could find a role in cancer therapy.
Collapse
Affiliation(s)
- Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| | - Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Ian F Hermans
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand; Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
7
|
Rollins MR, Spartz EJ, Stromnes IM. T Cell Receptor Engineered Lymphocytes for Cancer Therapy. ACTA ACUST UNITED AC 2020; 129:e97. [PMID: 32432843 DOI: 10.1002/cpim.97] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
T lymphocytes are capable of specific recognition and elimination of target cells. Physiological antigen recognition is mediated by the T cell receptor (TCR), which is an alpha beta heterodimer comprising the products of randomly rearranged V, D, and J genes. The exquisite specificity and functionality of T cells can be leveraged for cancer therapy: specifically, the adoptive transfer of T cells that express tumor-reactive TCRs can induce regression of solid tumors in patients with advanced cancer. However, the isolation and expression of a tumor antigen-specific TCRs is a highly involved process that requires identifying an immunogenic epitope, ensuring human cells are of the correct haplotype, performing a laborious T cell expansion process, and carrying out downstream TCR sequencing and cloning. Recent advances in single-cell sequencing have begun to streamline this process. This protocol synthesizes and expands upon methodologies to generate, isolate, and engineer human T cells with tumor-reactive TCRs for adoptive cell therapy. Though this process is perhaps more arduous than the alternative strategy of using chimeric antigen receptors (CARs) for engineering, the ability to target intracellular proteins using TCRs substantially increases the types of antigens that can be safely targeted. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Generation of human autologous dendritic cells from monocytes Basic Protocol 2: In vitro priming and expansion of human antigen-specific T cells Basic Protocol 3: Cloning of antigen-specific T cell receptors based on single-cell VDJ sequencing data Basic Protocol 4: Validation of T cell receptor expression and functionality in vitro Basic Protocol 5: Rapid expansion of T cell receptor-transduced T cells and human T cell clones.
Collapse
Affiliation(s)
- Meagan R Rollins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Ellen J Spartz
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Ingunn M Stromnes
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota.,Center for Genome Engineering, University of Minnesota Medical School, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
8
|
Bajwa G, Lanz I, Cardenas M, Brenner MK, Arber C. Transgenic CD8αβ co-receptor rescues endogenous TCR function in TCR-transgenic virus-specific T cells. J Immunother Cancer 2020; 8:e001487. [PMID: 33148692 PMCID: PMC7640589 DOI: 10.1136/jitc-2020-001487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Genetically engineered virus-specific T cells (VSTs) are a platform for adoptive cell therapy after allogeneic hematopoietic stem cell transplantation. However, redirection to a tumor-associated antigen by the introduction of a transgenic T-cell receptor (TCR) reduces anti-viral activity, thereby impeding the possibility of preventing or treating two distinct complications-malignant relapse and viral infection-with a single cell therapy product. Availability of CD8αβ co-receptor molecules can significantly impact class I restricted T-cell activation, and thus, we interrogated whether transgenic CD8αβ improves anti-viral activity mediated by native VSTs with or without a co-expressed transgenic TCR (TCR8). METHODS Our existing clinical VST manufacturing platform was adapted and validated to engineer TCR+ or TCR8+ VSTs targeting cytomegalovirus and Epstein-Barr virus. Simultaneous anti-viral and anti-tumor function of engineered VSTs was assessed in vitro and in vivo. We used pentamer staining, interferon (IFN)-γ enzyme-linked immunospot (ELISpot), intracellular cytokine staining (ICS), cytotoxicity assays, co-cultures, and cytokine secretion assays for the in vitro characterization. The in vivo anti-tumor function was assessed in a leukemia xenograft mouse model. RESULTS Both transgenic CD8αβ alone and TCR8 had significant impact on the anti-viral function of engineered VSTs, and TCR8+ VSTs had comparable anti-viral activity as non-engineered VSTs as determined by IFN-γ ELISpot, ICS and cytotoxicity assays. TCR8-engineered VSTs had improved anti-tumor function and greater effector cytokine production in vitro, as well as enhanced anti-tumor function against leukemia xenografts in mice. CONCLUSION Incorporation of transgenic CD8αβ into vectors for TCR-targetable antigens preserves anti-viral activity of TCR transgenic VSTs while simultaneously supporting tumor-directed activity mediated by a transgenic TCR. Our approach may provide clinical benefit in preventing and treating viral infections and malignant relapse post-transplant.
Collapse
Affiliation(s)
- Gagan Bajwa
- Department of Oncology UNIL CHUV, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Inès Lanz
- Department of Oncology UNIL CHUV, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Mara Cardenas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Caroline Arber
- Department of Oncology UNIL CHUV, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
9
|
Biernacki MA, Foster KA, Woodward KB, Coon ME, Cummings C, Cunningham TM, Dossa RG, Brault M, Stokke J, Olsen TM, Gardner K, Estey E, Meshinchi S, Rongvaux A, Bleakley M. CBFB-MYH11 fusion neoantigen enables T cell recognition and killing of acute myeloid leukemia. J Clin Invest 2020; 130:5127-5141. [PMID: 32831296 PMCID: PMC7524498 DOI: 10.1172/jci137723] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Proteins created from recurrent fusion genes like CBFB-MYH11 are prevalent in acute myeloid leukemia (AML), often necessary for leukemogenesis, persistent throughout the disease course, and highly leukemia specific, making them attractive neoantigen targets for immunotherapy. A nonameric peptide derived from a prevalent CBFB-MYH11 fusion protein was found to be immunogenic in HLA-B*40:01+ donors. High-avidity CD8+ T cell clones isolated from healthy donors killed CBFB-MYH11+ HLA-B*40:01+ AML cell lines and primary human AML samples in vitro. CBFB-MYH11-specific T cells also controlled CBFB-MYH11+ HLA-B*40:01+ AML in vivo in a patient-derived murine xenograft model. High-avidity CBFB-MYH11 epitope-specific T cell receptors (TCRs) transduced into CD8+ T cells conferred antileukemic activity in vitro. Our data indicate that the CBFB-MYH11 fusion neoantigen is naturally presented on AML blasts and enables T cell recognition and killing of AML. We provide proof of principle for immunologically targeting AML-initiating fusions and demonstrate that targeting neoantigens has clinical relevance even in low-mutational frequency cancers like fusion-driven AML. This work also represents a first critical step toward the development of TCR T cell immunotherapy targeting fusion gene-driven AML.
Collapse
Affiliation(s)
- Melinda A. Biernacki
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine
| | - Kimberly A. Foster
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kyle B. Woodward
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Michael E. Coon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Carrie Cummings
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Tanya M. Cunningham
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Robson G. Dossa
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Michelle Brault
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jamie Stokke
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pediatrics, and
| | - Tayla M. Olsen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Elihu Estey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pediatrics, and
| | - Anthony Rongvaux
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pediatrics, and
| |
Collapse
|
10
|
The Quest for the Best: How TCR Affinity, Avidity, and Functional Avidity Affect TCR-Engineered T-Cell Antitumor Responses. Cells 2020; 9:cells9071720. [PMID: 32708366 PMCID: PMC7408146 DOI: 10.3390/cells9071720] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Over the past decades, adoptive transfer of T cells has revolutionized cancer immunotherapy. In particular, T-cell receptor (TCR) engineering of T cells has marked important milestones in developing more precise and personalized cancer immunotherapies. However, to get the most benefit out of this approach, understanding the role that TCR affinity, avidity, and functional avidity play on how TCRs and T cells function in the context of tumor-associated antigen (TAA) recognition is vital to keep generating improved adoptive T-cell therapies. Aside from TCR-related parameters, other critical factors that govern T-cell activation are the effect of TCR co-receptors on TCR–peptide-major histocompatibility complex (pMHC) stabilization and TCR signaling, tumor epitope density, and TCR expression levels in TCR-engineered T cells. In this review, we describe the key aspects governing TCR specificity, T-cell activation, and how these concepts can be applied to cancer-specific TCR redirection of T cells.
Collapse
|
11
|
Luo XH, Meng Q, Liu Z, Paraschoudi G. Generation of high-affinity CMV-specific T cells for adoptive immunotherapy using IL-2, IL-15, and IL-21. Clin Immunol 2020; 217:108456. [PMID: 32376504 DOI: 10.1016/j.clim.2020.108456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Cytomegalovirus (CMV) infection remains a life-threatening condition in individuals with a suppressed immune system. CMV may also represent a clinically relevant target for immune responses in CMV-positive malignancies. We established a protocol to expand CMV-specific T cells (CMV-T) using peripheral blood mononuclear cells (PBMCs). PBMCs from 16 HLA-A*0201 donors were cultured with a cytokine cocktail comprising IL-2/IL-15/IL-21 along with overlapping peptides from CMV-pp65. Ten days later, T cells were stimulated with anti-CD3 (OKT3) and irradiated autologous PBMCs. CMV-T were detected by HLA-A*0201 CMV-pp65NLVPMVATV wild type and q226a mutant tetramers (for high-affinity T cells), intracellular cytokine staining, a CD107a mobilization assays as well as IFN-γ and TNF-α production in cell culture supernatants. We reliably obtained 50.25 ± 27.27% of CD8+ and 22.08 ± 21.83% of CD4+ T cells post-CMV-pp65 stimulation of PBMCs with a Th1-polarized phenotype and decreased Th2/Th17 responses. Most CD3 + CD8 + tetramer+ T cells were effector-memory cells, particularly among high-affinity CMV-T (q226a CMV-tetramer+). High-affinity CMV-T cells, compared to WT-tetramer+ cells, expressed higher IL-21R and lower FasL post-stimulation with CMV-pp65. The IL-2/IL-15/IL-21 cocktail also promoted CCR6 and CXCR3 expression necessary for T-cell migration into tissues. We have optimized methods for generating high-affinity CMV-specific T cells that can be used for adoptive cellular therapy in clinical practice.
Collapse
Affiliation(s)
- Xiao-Hua Luo
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing 400016, China.
| | - Qingda Meng
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zhenjiang Liu
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Georgia Paraschoudi
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Prota G, Gileadi U, Rei M, Lechuga-Vieco AV, Chen JL, Galiani S, Bedard M, Lau VWC, Fanchi LF, Artibani M, Hu Z, Gordon S, Rehwinkel J, Enríquez JA, Ahmed AA, Schumacher TN, Cerundolo V. Enhanced Immunogenicity of Mitochondrial-Localized Proteins in Cancer Cells. Cancer Immunol Res 2020; 8:685-697. [PMID: 32205315 DOI: 10.1158/2326-6066.cir-19-0467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 11/05/2019] [Accepted: 03/12/2020] [Indexed: 11/16/2022]
Abstract
Epitopes derived from mutated cancer proteins elicit strong antitumor T-cell responses that correlate with clinical efficacy in a proportion of patients. However, it remains unclear whether the subcellular localization of mutated proteins influences the efficiency of T-cell priming. To address this question, we compared the immunogenicity of NY-ESO-1 and OVA localized either in the cytosol or in mitochondria. We showed that tumors expressing mitochondrial-localized NY-ESO-1 and OVA proteins elicit significantdly higher frequencies of antigen-specific CD8+ T cells in vivo. We also demonstrated that this stronger immune response is dependent on the mitochondrial location of the antigenic proteins, which contributes to their higher steady-state amount, compared with cytosolic localized proteins. Consistent with these findings, we showed that injection of mitochondria purified from B16 melanoma cells can protect mice from a challenge with B16 cells, but not with irrelevant tumors. Finally, we extended these findings to cancer patients by demonstrating the presence of T-cell responses specific for mutated mitochondrial-localized proteins. These findings highlight the utility of prioritizing epitopes derived from mitochondrial-localized mutated proteins as targets for cancer vaccination strategies.
Collapse
Affiliation(s)
- Gennaro Prota
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Margarida Rei
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ana Victoria Lechuga-Vieco
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Ji-Li Chen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Silvia Galiani
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Melissa Bedard
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Vivian Wing Chong Lau
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Lorenzo F Fanchi
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mara Artibani
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom
| | - Zhiyuan Hu
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom
| | - Siamon Gordon
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
- Chang Gung University, Graduate Institute of Biomedical Sciences, College of Medicine, Taoyuan City, Taiwan
| | - Jan Rehwinkel
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Jose A Enríquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Ciber de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Ahmed A Ahmed
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom
| | - Ton N Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Abstract
T cells can be controllably stimulated through antigen-specific or nonspecific protocols. Accompanying functional hallmarks of T cell activation can include cytoskeletal reorganization, cell size increase, and cytokine secretion. Photon-induced near-field electron microscopy (PINEM) is used to image and quantify evanescent electric fields at the surface of T cells as a function of various stimulation conditions. While PINEM signal strength scales with multiple of the biophysical changes associated with T cell functional activation, it mostly strongly correlates with antigen-engagement of the T cell receptors, even under conditions that do not lead to functional T cell activation. PINEM image analysis suggests that a stimulation-induced reorganization of T cell surface structure, especially over length scales of a few hundred nanometers, is the dominant contributor to these PINEM signal changes. These experiments reveal that PINEM can provide a sensitive label-free probe of nanoscale cellular surface structures.
Collapse
|
14
|
Poiret T, Axelsson-Robertson R, Remberger M, Luo XH, Rao M, Nagchowdhury A, Von Landenberg A, Ernberg I, Ringden O, Maeurer M. Cytomegalovirus-Specific CD8+ T-Cells With Different T-Cell Receptor Affinities Segregate T-Cell Phenotypes and Correlate With Chronic Graft-Versus-Host Disease in Patients Post-Hematopoietic Stem Cell Transplantation. Front Immunol 2018; 9:760. [PMID: 29692783 PMCID: PMC5903031 DOI: 10.3389/fimmu.2018.00760] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 03/27/2018] [Indexed: 12/26/2022] Open
Abstract
Virus-specific T-cell responses are crucial to control cytomegalovirus (CMV) infections/reactivation in immunocompromised individuals. Adoptive cellular therapy with CMV-specific T-cells has become a viable treatment option. High-affinity anti-viral cellular immune responses are associated with improved long-term immune protection against CMV infection. To date, the characterization of high-affinity T-cell responses against CMV has not been achieved in blood from patients after allogeneic hematopoietic stem cell transplantation (HSCT). Therefore, the purpose of this study was to describe and analyze the phenotype and clinical impact of different CMV-specific CD8+ cytotoxic T-lymphocytes (CMV-CTL) classes based on their T-cell receptor (TCR) affinity. T-cells isolated from 23 patients during the first year following HSCT were tested for the expression of memory markers, programmed cell death 1 (PD-1), as well as TCR affinity, using three different HLA-A*02:01 CMVNLVPMVATV-Pp65 tetramers (wild-type, a245v and q226a mutants). High-affinity CMV-CTL defined by q226a tetramer binding, exhibited a higher frequency in CD8+ T-cells in the first month post-HSCT and exhibited an effector memory phenotype associated with strong PD-1 expression as compared to the medium- and low-affinity CMV-CTLs. High-affinity CMV-CTL was found at higher proportion in patients with chronic graft-versus-host disease (p < 0.001). This study provides a first insight into the detailed TCR affinities of CMV-CTL. This may be useful in order to improve current immunotherapy protocols using isolation of viral-specific T-cell populations based on their TCR affinity.
Collapse
Affiliation(s)
- Thomas Poiret
- Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Xiao-Hua Luo
- Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Rao
- Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Anurupa Nagchowdhury
- Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Von Landenberg
- Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Olle Ringden
- Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Markus Maeurer
- Department of Laboratory Medicine, Karolinska University Hospital, Stockholm, Sweden
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
Rius C, Attaf M, Tungatt K, Bianchi V, Legut M, Bovay A, Donia M, Thor Straten P, Peakman M, Svane IM, Ott S, Connor T, Szomolay B, Dolton G, Sewell AK. Peptide-MHC Class I Tetramers Can Fail To Detect Relevant Functional T Cell Clonotypes and Underestimate Antigen-Reactive T Cell Populations. THE JOURNAL OF IMMUNOLOGY 2018; 200:2263-2279. [PMID: 29483360 PMCID: PMC5857646 DOI: 10.4049/jimmunol.1700242] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 01/29/2018] [Indexed: 12/01/2022]
Abstract
Peptide-MHC (pMHC) multimers, usually used as streptavidin-based tetramers, have transformed the study of Ag-specific T cells by allowing direct detection, phenotyping, and enumeration within polyclonal T cell populations. These reagents are now a standard part of the immunology toolkit and have been used in many thousands of published studies. Unfortunately, the TCR-affinity threshold required for staining with standard pMHC multimer protocols is higher than that required for efficient T cell activation. This discrepancy makes it possible for pMHC multimer staining to miss fully functional T cells, especially where low-affinity TCRs predominate, such as in MHC class II–restricted responses or those directed against self-antigens. Several recent, somewhat alarming, reports indicate that pMHC staining might fail to detect the majority of functional T cells and have prompted suggestions that T cell immunology has become biased toward the type of cells amenable to detection with multimeric pMHC. We use several viral- and tumor-specific pMHC reagents to compare populations of human T cells stained by standard pMHC protocols and optimized protocols that we have developed. Our results confirm that optimized protocols recover greater populations of T cells that include fully functional T cell clonotypes that cannot be stained by regular pMHC-staining protocols. These results highlight the importance of using optimized procedures that include the use of protein kinase inhibitor and Ab cross-linking during staining to maximize the recovery of Ag-specific T cells and serve to further highlight that many previous quantifications of T cell responses with pMHC reagents are likely to have considerably underestimated the size of the relevant populations.
Collapse
Affiliation(s)
- Cristina Rius
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Meriem Attaf
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Katie Tungatt
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Valentina Bianchi
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Mateusz Legut
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Amandine Bovay
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom.,Department of Oncology and Ludwig Cancer Research, Lausanne University Hospital, Epalinges VD 1066, Switzerland
| | - Marco Donia
- Centre for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Per Thor Straten
- Centre for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Mark Peakman
- Department of Immunobiology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
| | - Inge Marie Svane
- Centre for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Sascha Ott
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Tom Connor
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom; and.,Cardiff University School of Biosciences, Cardiff CF10 3AX, United Kingdom
| | - Barbara Szomolay
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom
| | - Andrew K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom; .,Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital Wales, Cardiff CF14 4XN, United Kingdom; and
| |
Collapse
|
16
|
Cole DK, Fuller A, Dolton G, Zervoudi E, Legut M, Miles K, Blanchfield L, Madura F, Holland CJ, Bulek AM, Bridgeman JS, Miles JJ, Schauenburg AJA, Beck K, Evavold BD, Rizkallah PJ, Sewell AK. Dual Molecular Mechanisms Govern Escape at Immunodominant HLA A2-Restricted HIV Epitope. Front Immunol 2017; 8:1503. [PMID: 29209312 PMCID: PMC5701626 DOI: 10.3389/fimmu.2017.01503] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/25/2017] [Indexed: 12/05/2022] Open
Abstract
Serial accumulation of mutations to fixation in the SLYNTVATL (SL9) immunodominant, HIV p17 Gag-derived, HLA A2-restricted cytotoxic T lymphocyte epitope produce the SLFNTIAVL triple mutant “ultimate” escape variant. These mutations in solvent-exposed residues are believed to interfere with TCR recognition, although confirmation has awaited structural verification. Here, we solved a TCR co-complex structure with SL9 and the triple escape mutant to determine the mechanism of immune escape in this eminent system. We show that, in contrast to prevailing hypotheses, the main TCR contact residue is 4N and the dominant mechanism of escape is not via lack of TCR engagement. Instead, mutation of solvent-exposed residues in the peptide destabilise the peptide–HLA and reduce peptide density at the cell surface. These results highlight the extraordinary lengths that HIV employs to evade detection by high-affinity TCRs with a broad peptide-binding footprint and necessitate re-evaluation of this exemplar model of HIV TCR escape.
Collapse
Affiliation(s)
- David K Cole
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Anna Fuller
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Garry Dolton
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Efthalia Zervoudi
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Mateusz Legut
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Kim Miles
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Lori Blanchfield
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Florian Madura
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Christopher J Holland
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Anna M Bulek
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - John S Bridgeman
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - John J Miles
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom.,James Cook University, Cairns, QLD, Australia
| | - Andrea J A Schauenburg
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Konrad Beck
- Cardiff University School of Dentistry, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Brian D Evavold
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Pierre J Rizkallah
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| | - Andrew K Sewell
- Cardiff University School of Medicine, University Hospital, Heath Park, Cardiff, United Kingdom
| |
Collapse
|
17
|
Miller NJ, Church CD, Dong L, Crispin D, Fitzgibbon MP, Lachance K, Jing L, Shinohara M, Gavvovidis I, Willimsky G, McIntosh M, Blankenstein T, Koelle DM, Nghiem P. Tumor-Infiltrating Merkel Cell Polyomavirus-Specific T Cells Are Diverse and Associated with Improved Patient Survival. Cancer Immunol Res 2017; 5:137-147. [PMID: 28093446 PMCID: PMC5421625 DOI: 10.1158/2326-6066.cir-16-0210] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 01/02/2023]
Abstract
Tumor-infiltrating CD8+ T cells are associated with improved survival of patients with Merkel cell carcinoma (MCC), an aggressive skin cancer causally linked to Merkel cell polyomavirus (MCPyV). However, CD8+ T-cell infiltration is robust in only 4% to 18% of MCC tumors. We characterized the T-cell receptor (TCR) repertoire restricted to one prominent epitope of MCPyV (KLLEIAPNC, "KLL") and assessed whether TCR diversity, tumor infiltration, or T-cell avidity correlated with clinical outcome. HLA-A*02:01/KLL tetramer+ CD8+ T cells from MCC patient peripheral blood mononuclear cells (PBMC) and tumor-infiltrating lymphocytes (TIL) were isolated via flow cytometry. TCRβ (TRB) sequencing was performed on tetramer+ cells from PBMCs or TILs (n = 14) and matched tumors (n = 12). Functional avidity of T-cell clones was determined by IFNγ production. We identified KLL tetramer+ T cells in 14% of PBMC and 21% of TIL from MCC patients. TRB repertoires were strikingly diverse (397 unique TRBs were identified from 12 patients) and mostly private (only one TCRb clonotype shared between two patients). An increased fraction of KLL-specific TIL (>1.9%) was associated with significantly increased MCC-specific survival P = 0.0009). T-cell cloning from four patients identified 42 distinct KLL-specific TCRa/b pairs. T-cell clones from patients with improved MCC-specific outcomes were more avid (P < 0.05) and recognized an HLA-appropriate MCC cell line. T cells specific for a single MCPyV epitope display marked TCR diversity within and between patients. Intratumoral infiltration by MCPyV-specific T cells was associated with significantly improved MCC-specific survival, suggesting that augmenting the number or avidity of virus-specific T cells may have therapeutic benefit. Cancer Immunol Res; 5(2); 137-47. ©2017 AACR.
Collapse
MESH Headings
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Carcinoma, Merkel Cell/etiology
- Carcinoma, Merkel Cell/mortality
- Carcinoma, Merkel Cell/pathology
- Clonal Evolution/genetics
- Clonal Evolution/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Genetic Variation
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Merkel cell polyomavirus/immunology
- Prognosis
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Sequence Analysis, DNA
- Skin Neoplasms/etiology
- Skin Neoplasms/mortality
- Skin Neoplasms/pathology
- T-Cell Antigen Receptor Specificity/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Natalie J Miller
- Dermatology/Medicine/Pathology, University of Washington, Seattle, Washington
| | - Candice D Church
- Dermatology/Medicine/Pathology, University of Washington, Seattle, Washington
| | - Lichun Dong
- Department of Medicine/Laboratory Medicine/Global Health, University of Washington, Seattle, Washington
| | - David Crispin
- Fred Hutchinson, Public Health Sciences Division, Seattle, Washington
| | | | - Kristina Lachance
- Dermatology/Medicine/Pathology, University of Washington, Seattle, Washington
| | - Lichen Jing
- Department of Medicine/Laboratory Medicine/Global Health, University of Washington, Seattle, Washington
| | - Michi Shinohara
- Dermatology/Medicine/Pathology, University of Washington, Seattle, Washington
| | - Ioannis Gavvovidis
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Institute of Immunology, Charité, Berlin, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité, Berlin, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin McIntosh
- Fred Hutchinson, Public Health Sciences Division, Seattle, Washington
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Institute of Immunology, Charité, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - David M Koelle
- Department of Medicine/Laboratory Medicine/Global Health, University of Washington, Seattle, Washington.
- Fred Hutchinson, Vaccine and Infectious Disease Division, Seattle, Washington
- Benaroya Research Institute, Seattle, Washington
| | - Paul Nghiem
- Dermatology/Medicine/Pathology, University of Washington, Seattle, Washington.
| |
Collapse
|
18
|
Altman JD, Davis MM. MHC‐Peptide Tetramers to Visualize Antigen‐Specific T Cells. ACTA ACUST UNITED AC 2016; 115:17.3.1-17.3.44. [DOI: 10.1002/cpim.14] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Mark M. Davis
- Stanford University School of Medicine and The Howard Hughes Medical Institute Palo Alto California
| |
Collapse
|
19
|
Abstract
OBJECTIVES Strategies to cure HIV-1 infection require the eradication of viral reservoirs. An innovative approach for boosting the cytotoxic T-lymphocyte response is the transfer of T-cell receptors (TCRs). Previously, we have shown that electroporation of TCR-encoding mRNA is able to reprogram CD8 T cells derived from healthy donors. So far, it is unknown whether the transfer of HIV-1-specific TCRs is capable to reprogram CD8 T cells of HIV-1-infected patients. To assess the efficiency of TCR-transfer by mRNA electroporation and the functionality of reprogramed T cells in HIV-1-infected patients, we performed an in-vitro analysis of TCR-transfer into T cells from HIV-1-infected patients in various stages of disease and from healthy controls. METHODS Peripheral blood mononuclear cells from 16 HIV-1-infected patients (nine HLA-A02-positive, seven HLA-A02-negative) and from five healthy controls were electroporated with mRNA-constructs encoding TCRs specific for the HLA-A02/HIV-1-gag p17 epitope SLYNTVATL (SL9). Functionality of the TCRs was measured by γIFN-ELISpot assays. RESULTS SL9/TCR transfection into peripheral blood mononuclear cells from both HLA-A02-positive and HLA-A02-negative HIV-1-infected patients and from healthy blood donors reprogramed T cells for recognition of SL9-presenting HLA-A02-positive cells in γIFN-ELISpot assays. SL9/TCR-transfer into T cells from an immunodeficient AIDS patient could induce recognition of SL9-expressing target cells only after reversion of T-cell dysfunction by antiretroviral therapy. CONCLUSION The transfer of HIV-1-p17-specific TCRs into T cells is functional both in HIV-1-infected patients as well as in healthy blood donors. TCR-transfer is a promising method to boost the immune system against HIV-1.
Collapse
|
20
|
Holland CJ, Dolton G, Scurr M, Ladell K, Schauenburg AJ, Miners K, Madura F, Sewell AK, Price DA, Cole DK, Godkin AJ. Enhanced Detection of Antigen-Specific CD4+ T Cells Using Altered Peptide Flanking Residue Peptide-MHC Class II Multimers. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:5827-36. [PMID: 26553072 PMCID: PMC4671089 DOI: 10.4049/jimmunol.1402787] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 10/08/2015] [Indexed: 11/22/2022]
Abstract
Fluorochrome-conjugated peptide-MHC (pMHC) class I multimers are staple components of the immunologist's toolbox, enabling reliable quantification and analysis of Ag-specific CD8(+) T cells irrespective of functional outputs. In contrast, widespread use of the equivalent pMHC class II (pMHC-II) reagents has been hindered by intrinsically weaker TCR affinities for pMHC-II, a lack of cooperative binding between the TCR and CD4 coreceptor, and a low frequency of Ag-specific CD4(+) T cell populations in the peripheral blood. In this study, we show that peptide flanking regions, extending beyond the central nonamer core of MHC-II-bound peptides, can enhance TCR-pMHC-II binding and T cell activation without loss of specificity. Consistent with these findings, pMHC-II multimers incorporating peptide flanking residue modifications proved superior for the ex vivo detection, characterization, and manipulation of Ag-specific CD4(+) T cells, highlighting an unappreciated feature of TCR-pMHC-II interactions.
Collapse
Affiliation(s)
- Christopher J Holland
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Martin Scurr
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Andrea J Schauenburg
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Kelly Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Florian Madura
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Andrew K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - David K Cole
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and
| | - Andrew J Godkin
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, Wales, United Kingdom; and Department of Integrated Medicine, University Hospital of Wales, Cardiff CF14 4XW, Wales, United Kingdom
| |
Collapse
|
21
|
Dolton G, Tungatt K, Lloyd A, Bianchi V, Theaker SM, Trimby A, Holland CJ, Donia M, Godkin AJ, Cole DK, Straten PT, Peakman M, Svane IM, Sewell AK. More tricks with tetramers: a practical guide to staining T cells with peptide-MHC multimers. Immunology 2015; 146:11-22. [PMID: 26076649 PMCID: PMC4552497 DOI: 10.1111/imm.12499] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 05/27/2015] [Indexed: 12/15/2022] Open
Abstract
Analysis of antigen-specific T-cell populations by flow cytometry with peptide-MHC (pMHC) multimers is now commonplace. These reagents allow the tracking and phenotyping of T cells during infection, autoimmunity and cancer, and can be particularly revealing when used for monitoring therapeutic interventions. In 2009, we reviewed a number of 'tricks' that could be used to improve this powerful technology. More recent advances have demonstrated the potential benefits of using higher order multimers and of 'boosting' staining by inclusion of an antibody against the pMHC multimer. These developments now allow staining of T cells where the interaction between the pMHC and the T-cell receptor is over 20-fold weaker (K(D) > 1 mm) than could previously be achieved. Such improvements are particularly relevant when using pMHC multimers to stain anti-cancer or autoimmune T-cell populations, which tend to bear lower affinity T-cell receptors. Here, we update our previous work to include discussion of newer tricks that can produce substantially brighter staining even when using log-fold lower concentrations of pMHC multimer. We further provide a practical guide to using pMHC multimers that includes a description of several common pitfalls and how to circumvent them.
Collapse
Affiliation(s)
- Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Katie Tungatt
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Angharad Lloyd
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Valentina Bianchi
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Sarah M Theaker
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Andrew Trimby
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Christopher J Holland
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Marco Donia
- Centre for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | - Andrew J Godkin
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - David K Cole
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Per Thor Straten
- Centre for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | - Mark Peakman
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital, London, UK
| | - Inge Marie Svane
- Centre for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | - Andrew K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
22
|
Tungatt K, Bianchi V, Crowther MD, Powell WE, Schauenburg AJ, Trimby A, Donia M, Miles JJ, Holland CJ, Cole DK, Godkin AJ, Peakman M, Straten PT, Svane IM, Sewell AK, Dolton G. Antibody stabilization of peptide-MHC multimers reveals functional T cells bearing extremely low-affinity TCRs. THE JOURNAL OF IMMUNOLOGY 2014; 194:463-74. [PMID: 25452566 PMCID: PMC4273996 DOI: 10.4049/jimmunol.1401785] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fluorochrome-conjugated peptide–MHC (pMHC) multimers are commonly used in combination with flow cytometry for direct ex vivo visualization and characterization of Ag-specific T cells, but these reagents can fail to stain cells when TCR affinity and/or TCR cell-surface density are low. pMHC multimer staining of tumor-specific, autoimmune, or MHC class II–restricted T cells can be particularly challenging, as these T cells tend to express relatively low-affinity TCRs. In this study, we attempted to improve staining using anti-fluorochrome unconjugated primary Abs followed by secondary staining with anti-Ab fluorochrome-conjugated Abs to amplify fluorescence intensity. Unexpectedly, we found that the simple addition of an anti-fluorochrome unconjugated Ab during staining resulted in considerably improved fluorescence intensity with both pMHC tetramers and dextramers and with PE-, allophycocyanin-, or FITC-based reagents. Importantly, when combined with protein kinase inhibitor treatment, Ab stabilization allowed pMHC tetramer staining of T cells even when the cognate TCR–pMHC affinity was extremely low (KD >1 mM) and produced the best results that we have observed to date. We find that this inexpensive addition to pMHC multimer staining protocols also allows improved recovery of cells that have recently been exposed to Ag, improvements in the recovery of self-specific T cells from PBMCs or whole-blood samples, and the use of less reagent during staining. In summary, Ab stabilization of pMHC multimers during T cell staining extends the range of TCR affinities that can be detected, yields considerably enhanced staining intensities, and is compatible with using reduced amounts of these expensive reagents.
Collapse
Affiliation(s)
- Katie Tungatt
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Valentina Bianchi
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Michael D Crowther
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Wendy E Powell
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Andrea J Schauenburg
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Andrew Trimby
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Marco Donia
- Center for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - John J Miles
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom; QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia; and
| | - Christopher J Holland
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - David K Cole
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Andrew J Godkin
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| | - Mark Peakman
- Department of Immunobiology, King's College London School of Medicine, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Per Thor Straten
- Center for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Andrew K Sewell
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom;
| | - Garry Dolton
- Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, Wales, United Kingdom
| |
Collapse
|
23
|
Amino acids and immune response: a role for cysteine, glutamine, phenylalanine, tryptophan and arginine in T-cell function and cancer? Pathol Oncol Res 2014; 21:9-17. [PMID: 25351939 DOI: 10.1007/s12253-014-9860-0] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 10/22/2014] [Indexed: 01/16/2023]
Abstract
While proteins are critical for immunity, T-cells constitute a critical component of adaptive immunity by clearing cancerous cells among other abnormal cells. However, cancer cells exhibit a potential to escape T-cell control by employing mechanisms not completely delineated. Interesting work has investigated how certain amino acids affect the proliferation rate of T-cells as well as their effectiveness in clearing tumors. The role of amino acids cysteine, glutamine, phenylalanine, tryptophan and arginine in immunomodulation and particularly regarding T-cell proliferation and activation is discussed. The redox balance is reported to affect T-cell proliferation via modulation of cysteine availability. In addition antigen presenting cells (APCs), similar to myeloid cells determine the availability of amino acids in the extracellular microenvironment affecting T-cell proliferation and activation. A better mechanistic understanding of T-cell function modulation via amino acid signaling or metabolic properties may be helpful towards optimization of adaptive immunity with implications for cancer prognosis and treatment.
Collapse
|
24
|
Dolton G, Lissina A, Skowera A, Ladell K, Tungatt K, Jones E, Kronenberg-Versteeg D, Akpovwa H, Pentier JM, Holland CJ, Godkin AJ, Cole DK, Neller MA, Miles JJ, Price DA, Peakman M, Sewell AK. Comparison of peptide-major histocompatibility complex tetramers and dextramers for the identification of antigen-specific T cells. Clin Exp Immunol 2014; 177:47-63. [PMID: 24673376 PMCID: PMC4089154 DOI: 10.1111/cei.12339] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2014] [Indexed: 02/05/2023] Open
Abstract
Fluorochrome-conjugated peptide-major histocompatibility complex (pMHC) multimers are widely used for flow cytometric visualization of antigen-specific T cells. The most common multimers, streptavidin-biotin-based 'tetramers', can be manufactured readily in the laboratory. Unfortunately, there are large differences between the threshold of T cell receptor (TCR) affinity required to capture pMHC tetramers from solution and that which is required for T cell activation. This disparity means that tetramers sometimes fail to stain antigen-specific T cells within a sample, an issue that is particularly problematic when staining tumour-specific, autoimmune or MHC class II-restricted T cells, which often display TCRs of low affinity for pMHC. Here, we compared optimized staining with tetramers and dextramers (dextran-based multimers), with the latter carrying greater numbers of both pMHC and fluorochrome per molecule. Most notably, we find that: (i) dextramers stain more brightly than tetramers; (ii) dextramers outperform tetramers when TCR-pMHC affinity is low; (iii) dextramers outperform tetramers with pMHC class II reagents where there is an absence of co-receptor stabilization; and (iv) dextramer sensitivity is enhanced further by specific protein kinase inhibition. Dextramers are compatible with current state-of-the-art flow cytometry platforms and will probably find particular utility in the fields of autoimmunity and cancer immunology.
Collapse
Affiliation(s)
- G Dolton
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cukalac T, Chadderton J, Zeng W, Cullen JG, Kan WT, Doherty PC, Jackson DC, Turner SJ, La Gruta NL. The Influenza Virus–Specific CTL Immunodominance Hierarchy in Mice Is Determined by the Relative Frequency of High-Avidity T Cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:4061-8. [DOI: 10.4049/jimmunol.1301403] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
26
|
Combinatorial HLA-peptide bead libraries for high throughput identification of CD8⁺ T cell specificity. J Immunol Methods 2013; 403:72-8. [PMID: 24309405 DOI: 10.1016/j.jim.2013.11.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/18/2013] [Accepted: 11/20/2013] [Indexed: 11/23/2022]
Abstract
Comprehensive antigenic characterization of a T cell population of unknown specificity is challenging. Existing MHC class I expression systems are limited by the practical difficulty of probing cell populations with an MHC class I peptide library and the cross-reactivity of T cells that are able to recognise many variants of an index peptide. Using emulsion PCR and emulsion in vitro transcription/translation of a random library of peptides conjugated to CD8-null HLA-A*0201 on beads, we probed HLA-A*0201-restricted T cells with specificity for influenza, CMV and EBV. We observed significant enrichment for sequences containing HLA-A2 anchors and correct viral fragments for all T cell populations. HLA bead display provides a novel approach to identify the specificity of T cells.
Collapse
|
27
|
Characterization of T-cell receptors directed against HLA-A*01-restricted and C*07-restricted epitopes of MAGE-A3 and MAGE-A12. J Immunother 2013; 35:680-8. [PMID: 23090077 DOI: 10.1097/cji.0b013e31827338ea] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The ability of T cells that have been genetically engineered to express T-cell receptors (TCRs) directed against tumor antigens to mediate tumor regression has been demonstrated in several clinical trials. These TCRs have primarily targeted HLA-A*0201-restricted TCRs, as approximately 50% of whites, who represent the predominant population of patients who develop melanomas, expresses this HLA class I allele. These therapies could be extended to additional patients through the use of TCRs that target epitopes that are presented by additional class I alleles that are prevalent in this population such as HLA-C*07 and HLA-A*01, which are expressed by approximately 50% and 30% of the patient population respectively. Therefore, 2 TCRs that recognize an epitope of MAGE-A12 in the context of HLA-C*07 and 2 TCRs that recognize an epitope of MAGE-A3 in the context of HLA-A*01 were isolated from tumor-reactive T-cell clones and cloned in a recombinant retroviral expression vector. Comparative studies indicated that one of the 2 MAGE-A3-reactive TCRs and one of the 2 MAGE-A12-reactive TCRs were superior to the additional TCRs in conferring transduced peripheral blood mononuclear cells with the capacity to recognize a broad array of antigen and MHC-positive target cells. These results provide support for the use of these TCRs in cancer adoptive immunotherapy trials.
Collapse
|
28
|
Malecek K, Zhong S, McGary K, Yu C, Huang K, Johnson LA, Rosenberg SA, Krogsgaard M. Engineering improved T cell receptors using an alanine-scan guided T cell display selection system. J Immunol Methods 2013; 392:1-11. [PMID: 23500145 DOI: 10.1016/j.jim.2013.02.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/28/2013] [Accepted: 02/28/2013] [Indexed: 11/18/2022]
Abstract
T cell receptors (TCRs) on T cells recognize peptide-major histocompatibility complex (pMHC) molecules on the surface of antigen presenting cells and this interaction determines the T cell immune response. Due to negative selection, naturally occurring TCRs bind self (tumor) peptides with low affinity and have a much higher affinity for foreign antigens. This complicates isolation of naturally occurring, high affinity TCRs that mediate more effective tumor rejection for therapeutic purposes. An attractive approach to resolve this issue is to engineer high affinity TCRs in vitro using phage, yeast or mammalian TCR display systems. A caveat of these systems is that they rely on a large library by random mutagenesis due to the lack of knowledge regarding the specific interactions between the TCR and pMHC. We have focused on the mammalian retroviral display system because it uniquely allows for direct comparison of TCR-pMHC-binding properties with T-cell activation outcomes. Through an alanine-scanning approach, we are able to quickly map the key amino acid residues directly involved in TCR-pMHC interactions thereby significantly reducing the library size. Using this method, we demonstrate that for a self-antigen-specific human TCR (R6C12) the key residues for pMHC binding are located in the CDR3β region. This information was used as a basis for designing an efficacious TCR CDR3α library that allowed for selection of TCRs with higher avidity than the wild-type as evaluated through binding and activation experiments. This is a direct approach to target specific TCR residues in TCR library design to efficiently engineer high avidity TCRs that may potentially be used to enhance adoptive immunotherapy treatments.
Collapse
Affiliation(s)
- Karolina Malecek
- NYU Cancer institute, New York University School of Medicine, NewYork, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Functional avidity: a measure to predict the efficacy of effector T cells? Clin Dev Immunol 2012; 2012:153863. [PMID: 23227083 PMCID: PMC3511839 DOI: 10.1155/2012/153863] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 10/22/2012] [Indexed: 01/30/2023]
Abstract
The functional avidity is determined by exposing T-cell populations in vitro to different amounts of cognate antigen. T-cells with high functional avidity respond to low antigen doses. This in vitro measure is thought to correlate well with the in vivo effector capacity of T-cells. We here present the multifaceted factors determining and influencing the functional avidity of T-cells. We outline how changes in the functional avidity can occur over the course of an infection. This process, known as avidity maturation, can occur despite the fact that T-cells express a fixed TCR. Furthermore, examples are provided illustrating the importance of generating T-cell populations that exhibit a high functional avidity when responding to an infection or tumors. Furthermore, we discuss whether criteria based on which we evaluate an effective T-cell response to acute infections can also be applied to chronic infections such as HIV. Finally, we also focus on observations that high-avidity T-cells show higher signs of exhaustion and facilitate the emergence of virus escape variants. The review summarizes our current understanding of how this may occur as well as how T-cells of different functional avidity contribute to antiviral and anti-tumor immunity. Enhancing our knowledge in this field is relevant for tumor immunotherapy and vaccines design.
Collapse
|
30
|
Legoux FP, Moon JJ. Peptide:MHC tetramer-based enrichment of epitope-specific T cells. J Vis Exp 2012:4420. [PMID: 23117190 DOI: 10.3791/4420] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
A basic necessity for researchers studying adaptive immunity with in vivo experimental models is an ability to identify T cells based on their T cell antigen receptor (TCR) specificity. Many indirect methods are available in which a bulk population of T cells is stimulated in vitro with a specific antigen and epitope-specific T cells are identified through the measurement of a functional response such as proliferation, cytokine production, or expression of activation markers(1). However, these methods only identify epitope-specific T cells exhibiting one of many possible functions, and they are not sensitive enough to detect epitope-specific T cells at naive precursor frequencies. A popular alternative is the TCR transgenic adoptive transfer model, in which monoclonal T cells from a TCR transgenic mouse are seeded into histocompatible hosts to create a large precursor population of epitope-specific T cells that can be easily tracked with the use of a congenic marker antibody(2,3). While powerful, this method suffers from experimental artifacts associated with the unphysiological frequency of T cells with specificity for a single epitope(4,5). Moreover, this system cannot be used to investigate the functional heterogeneity of epitope-specific T cell clones within a polyclonal population. The ideal way to study adaptive immunity should involve the direct detection of epitope-specific T cells from the endogenous T cell repertoire using a method that distinguishes TCR specificity solely by its binding to cognate peptide:MHC (pMHC) complexes. The use of pMHC tetramers and flow cytometry accomplishes this(6), but is limited to the detection of high frequency populations of epitope-specific T cells only found following antigen-induced clonal expansion. In this protocol, we describe a method that coordinates the use of pMHC tetramers and magnetic cell enrichment technology to enable detection of extremely low frequency epitope-specific T cells from mouse lymphoid tissues(3,7). With this technique, one can comprehensively track entire epitope-specific populations of endogenous T cells in mice at all stages of the immune response.
Collapse
Affiliation(s)
- Francois P Legoux
- Center for Immunology and Inflammatory Diseases, and Pulmonary and Critical Care Unit, Massachusetts General Hospital and Harvard Medical School, USA
| | | |
Collapse
|
31
|
Suzuki S, Masaki A, Ishida T, Ito A, Mori F, Sato F, Narita T, Ri M, Kusumoto S, Komatsu H, Fukumori Y, Nishikawa H, Tanaka Y, Niimi A, Inagaki H, Iida S, Ueda R. Tax is a potential molecular target for immunotherapy of adult T-cell leukemia/lymphoma. Cancer Sci 2012; 103:1764-73. [PMID: 22735080 DOI: 10.1111/j.1349-7006.2012.02371.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 06/18/2012] [Accepted: 06/21/2012] [Indexed: 11/30/2022] Open
Abstract
We expanded CTL specific for Tax (a human T-lymphotropic virus type-1-encoded gene product) in vitro from PBMC of several adult T-cell leukemia/lymphoma (ATL) patients, and document its potential significance as a target for ATL immunotherapy. Tax-specific CTL responses against tumor cells were restricted by Tax-expression and the appropriate human leukocyte antigen (HLA) type. Tax-specific CTL recognized HLA/Tax-peptide complexes on autologous ATL cells, even when their Tax expression was so low that it could only be detected by RT-PCR but not by flow cytometry. Recognition resulted in interferon gamma (IFN-γ) production and target cell lysis. This would be the first report that Tax-specific CTL from ATL patients specifically recognized and killed autologous tumor cells that expressed Tax. The Tax-specific CTL responded to as little as 0.01 pM of the corresponding peptide, indicating that their T-cell receptor avidity was much higher than that of any other CTL recognizing viral or other tumor antigens. This is presumably the reason why the Tax-specific CTL recognized and killed autologous ATL cells despite their very low Tax expression. In addition, cell cycle analyses and experiments with primary ATL cell-bearing mice demonstrated that ATL cells present at the site of active cell proliferation, such as in the tumor masses, expressed substantial amounts of Tax, but it was minimally expressed by the tumor cells in a quiescent state, such as in the blood. The present study not only provides a strong rationale for exploiting Tax as a possible target for ATL immunotherapy but also contributes to our understanding of the immunopathogenesis of ATL.
Collapse
Affiliation(s)
- Susumu Suzuki
- Department of Medical Oncology & Immunology, Nagoya City University Graduate School of Medical Sciences, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
TCR gene transfer: MAGE-C2/HLA-A2 and MAGE-A3/HLA-DP4 epitopes as melanoma-specific immune targets. Clin Dev Immunol 2012; 2012:586314. [PMID: 22400038 PMCID: PMC3287115 DOI: 10.1155/2012/586314] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 10/07/2011] [Accepted: 10/20/2011] [Indexed: 01/03/2023]
Abstract
Adoptive therapy with TCR gene-engineered T cells provides an attractive and feasible treatment option for cancer patients. Further development of TCR gene therapy requires the implementation of T-cell target epitopes that prevent “on-target” reactivity towards healthy tissues and at the same time direct a clinically effective response towards tumor tissues. Candidate epitopes that meet these criteria are MAGE-C2336-344/HLA-A2 (MC2/A2) and MAGE-A3243-258/HLA-DP4 (MA3/DP4). We molecularly characterized TCRαβ genes of an MC2/A2-specific CD8 and MA3/DP4-specific CD4 T-cell clone derived from melanoma patients who responded clinically to MAGE vaccination. We identified MC2/A2 and MA3/DP4-specific TCR-Vα3/Vβ28 and TCR-Vα38/Vβ2 chains and validated these TCRs in vitro upon gene transfer into primary human T cells. The MC2 and MA3 TCR were surface-expressed and mediated CD8 T-cell functions towards melanoma cell lines and CD4 T-cell functions towards dendritic cells, respectively. We intend to start testing these MAGE-specific TCRs in phase I clinical trial.
Collapse
|
33
|
Imataki O, Ansén S, Tanaka M, Butler MO, Berezovskaya A, Milstein MI, Kuzushima K, Nadler LM, Hirano N. IL-21 can supplement suboptimal Lck-independent MAPK activation in a STAT-3-dependent manner in human CD8(+) T cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:1609-19. [PMID: 22238455 DOI: 10.4049/jimmunol.1003446] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although both MHC class II/CD8α double-knockout and CD8β null mice show a defect in the development of MHC class I-restricted CD8(+) T cells in the thymus, they possess low numbers of high-avidity peripheral CTL with limited clonality and are able to contain acute and chronic infections. These in vivo data suggest that the CD8 coreceptor is not absolutely necessary for the generation of Ag-specific CTL. Lack of CD8 association causes partial TCR signaling because of the absence of CD8/Lck recruitment to the proximity of the MHC/TCR complex, resulting in suboptimal MAPK activation. Therefore, there should exist a signaling mechanism that can supplement partial TCR activation caused by the lack of CD8 association. In this human study, we have shown that CD8-independent stimulation of Ag-specific CTL previously primed in the presence of CD8 coligation, either in vivo or in vitro, induced severely impaired in vitro proliferation. When naive CD8(+) T cells were primed in the absence of CD8 binding and subsequently restimulated in the presence of CD8 coligation, the proliferation of Ag-specific CTL was also severely hampered. However, when CD8-independent T cell priming and restimulation were supplemented with IL-21, Ag-specific CD8(+) CTL expanded in two of six individuals tested. We found that IL-21 rescued partial MAPK activation in a STAT3- but not STAT1-dependent manner. These results suggest that CD8 coligation is critical for the expansion of postthymic peripheral Ag-specific CTL in humans. However, STAT3-mediated IL-21 signaling can supplement partial TCR signaling caused by the lack of CD8 association.
Collapse
Affiliation(s)
- Osamu Imataki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Davis MM, Altman JD, Newell EW. Interrogating the repertoire: broadening the scope of peptide-MHC multimer analysis. Nat Rev Immunol 2011; 11:551-8. [PMID: 21760610 PMCID: PMC3699324 DOI: 10.1038/nri3020] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Labelling antigen-specific T cells with peptide-MHC multimers has provided an invaluable way to monitor T cell-mediated immune responses. A number of recent developments in this technology have made these multimers much easier to make and use in large numbers. Furthermore, enrichment techniques have provided a greatly increased sensitivity that allows the analysis of the naive T cell repertoire directly. Thus, we can expect a flood of new information to emerge in the coming years.
Collapse
Affiliation(s)
- Mark M Davis
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA.
| | | | | |
Collapse
|
35
|
Kitazono T, Okazaki T, Araya N, Yamano Y, Yamada Y, Nakamura T, Tanaka Y, Inoue M, Ozaki S. Advantage of higher-avidity CTL specific for Tax against human T-lymphotropic virus-1 infected cells and tumors. Cell Immunol 2011; 272:11-7. [DOI: 10.1016/j.cellimm.2011.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/06/2011] [Accepted: 10/03/2011] [Indexed: 01/30/2023]
|
36
|
Legoux F, Debeaupuis E, Echasserieau K, De La Salle H, Saulquin X, Bonneville M. Impact of TCR reactivity and HLA phenotype on naive CD8 T cell frequency in humans. THE JOURNAL OF IMMUNOLOGY 2010; 184:6731-8. [PMID: 20483723 DOI: 10.4049/jimmunol.1000295] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The impact of MHC phenotype on the shaping of the peripheral naive T cell repertoire in humans remains unknown. To address this, we compared the frequency and antigenic avidity of naive T cells specific for immunodominant self-, viral, and tumor Ags presented by a human MHC class I allele (HLA-A*02, referred to as A2) in individuals expressing or not this allele. Naive T cell frequencies varied from one Ag specificity to another but were restrained for a given specificity. Although A2-restricted T cells showed similar repertoire features and antigenic avidities in A2+ and A2- donors, A2 expression had either a positive, neutral, or negative impact on the frequency of A2-restricted naive CD8 T cells, depending on their fine specificity. We also identified in all donors CD4 T cells specific for A2/peptide complexes, whose frequencies were not affected by MHC class I expression, but nevertheless correlated with those of their naive CD8 T cell counterparts. Therefore, both selection by self-MHC and inherent TCR reactivity regulate the frequency of human naive T cell precursors. Moreover this study also suggests that T cell repertoire shaping by a given self-MHC allele is dispensable for generation of immunodominant T cell responses restricted by this particular allele.
Collapse
Affiliation(s)
- François Legoux
- Institut National de la Santé et de la Recherche Médicale Unité 892, Université de Nantes, Nantes, France
| | | | | | | | | | | |
Collapse
|
37
|
Oral vaccination with attenuated Salmonella enterica strains encoding T-cell epitopes from tumor antigen NY-ESO-1 induces specific cytotoxic T-lymphocyte responses. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:889-94. [PMID: 20375244 DOI: 10.1128/cvi.00044-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bacterial fimbriae can accept foreign peptides and display them on the cell surface. A highly efficient gene replacement method was used to generate peptide vaccines based on Salmonella enterica serovar Typhimurium SL3261. The T-cell epitopes (NY-ESO-1 p157-165 and p157-167) from NY-ESO-1, which is a promising target antigen in patients for the specific immune recognition of cancer, were incorporated into the gene encoding AgfA (the major subunit protein of thin aggregative fimbriae of Salmonella) by replacing an equal length of the DNA segment. To improve cytotoxic T-lymphocyte recognition, both termini of the peptide were flanked by double alanine (AA) residues. Immunofluorescence microscopy with AgfA-specific antiserum verified the expression of chimeric AgfA, which was also proved by a Congo red binding assay. Oral immunizations of HLA-A*0201 transgenic mice with recombinant SL3261 strains encoding NY-ESO-1 p157-165 or p157-167 induced NY-ESO-1 p157-165-specific CD8(+) T cells, detected by an HLA-A*0201 pentamer, and induced a T-cell response detected by an enzyme-linked immunospot assay. The Salmonella fimbrial display system was efficient at the induction of an antitumor cellular immune response in vivo, providing a new strategy for the development of efficient cancer vaccinations.
Collapse
|
38
|
Wooldridge L, Clement M, Lissina A, Edwards ESJ, Ladell K, Ekeruche J, Hewitt RE, Laugel B, Gostick E, Cole DK, Debets R, Berrevoets C, Miles JJ, Burrows SR, Price DA, Sewell AK. MHC class I molecules with Superenhanced CD8 binding properties bypass the requirement for cognate TCR recognition and nonspecifically activate CTLs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:3357-66. [PMID: 20190139 PMCID: PMC3024536 DOI: 10.4049/jimmunol.0902398] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD8(+) CTLs are essential for effective immune defense against intracellular microbes and neoplasia. CTLs recognize short peptide fragments presented in association with MHC class I (MHCI) molecules on the surface of infected or dysregulated cells. Ag recognition involves the binding of both TCR and CD8 coreceptor to a single ligand (peptide MHCI [pMHCI]). The TCR/pMHCI interaction confers Ag specificity, whereas the pMHCI/CD8 interaction mediates enhanced sensitivity to Ag. Striking biophysical differences exist between the TCR/pMHCI and pMHCI/CD8 interactions; indeed, the pMHCI/CD8 interaction can be >100-fold weaker than the cognate TCR/pMHCI interaction. In this study, we show that increasing the strength of the pMHCI/CD8 interaction by approximately 15-fold results in nonspecific, cognate Ag-independent pMHCI tetramer binding at the cell surface. Furthermore, pMHCI molecules with superenhanced affinity for CD8 activate CTLs in the absence of a specific TCR/pMHCI interaction to elicit a full range of effector functions, including cytokine/chemokine release, degranulation and proliferation. Thus, the low solution binding affinity of the pMHCI/CD8 interaction is essential for the maintenance of CTL Ag specificity.
Collapse
Affiliation(s)
- Linda Wooldridge
- Department of Infection, Cardiff University, Cardiff, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chen JL, Morgan AJ, Stewart-Jones G, Shepherd D, Bossi G, Wooldridge L, Hutchinson SL, Sewell AK, Griffiths GM, van der Merwe PA, Jones EY, Galione A, Cerundolo V. Ca2+ release from the endoplasmic reticulum of NY-ESO-1-specific T cells is modulated by the affinity of TCR and by the use of the CD8 coreceptor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:1829-1839. [PMID: 20053942 PMCID: PMC4222200 DOI: 10.4049/jimmunol.0902103] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Although several cancer immunotherapy strategies are based on the use of analog peptides and on the modulation of the TCR affinity of adoptively transferred T cells, it remains unclear whether tumor-specific T cell activation by strong and weak TCR stimuli evoke different Ca(2+) signatures from the Ca(2+) intracellular stores and whether the amplitude of Ca(2+) release from the endoplasmic reticulum (ER) can be further modulated by coreceptor binding to peptide/MHC. In this study, we combined functional, structural, and kinetic measurements to correlate the intensity of Ca(2+) signals triggered by the stimulation of the 1G4 T cell clone specific to the tumor epitope NY-ESO-1(157-165). Two analogs of the NY-ESO-1(157-165) peptide, having similar affinity to HLA-A2 molecules, but a 6-fold difference in binding affinity for the 1G4 TCR, resulted in different Ca(2+) signals and T cell activation. 1G4 stimulation by the stronger stimulus emptied the ER of stored Ca(2+), even in the absence of CD8 binding, resulting in sustained Ca(2+) influx. In contrast, the weaker stimulus induced only partial emptying of stored Ca(2+), resulting in significantly diminished and oscillatory Ca(2+) signals, which were enhanced by CD8 binding. Our data define the range of TCR/peptide MHC affinities required to induce depletion of Ca(2+) from intracellular stores and provide insights into the ability of T cells to tailor the use of the CD8 coreceptor to enhance Ca(2+) release from the ER. This, in turn, modulates Ca(2+) influx from the extracellular environment, ultimately controlling T cell activation.
Collapse
Affiliation(s)
- Ji-Li Chen
- Weatherall Institute of Molecular Medicine, OX3 9DS, Oxford
| | - Anthony J. Morgan
- Dept of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Guillaume Stewart-Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Dawn Shepherd
- Weatherall Institute of Molecular Medicine, OX3 9DS, Oxford
| | - Giovanna Bossi
- Immunocore, Limited, 57c Milton Park, Abingdon, Oxon, OX14 4RX, UK
| | - Linda Wooldridge
- Dept of Infection, Immunity and Biochemistry, The Henry Wellcome Building, Cardiff University School of Medicine, Cardiff, CF14 4XN
| | | | - Andrew K. Sewell
- Dept of Infection, Immunity and Biochemistry, The Henry Wellcome Building, Cardiff University School of Medicine, Cardiff, CF14 4XN
| | - Gillian M. Griffiths
- Cambridge Institute for Medical Research, PO Box 139, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0XY, UK
| | | | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Antony Galione
- Dept of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | | |
Collapse
|
40
|
Casalegno-Garduño R, Schmitt A, Yao J, Wang X, Xu X, Freund M, Schmitt M. Multimer technologies for detection and adoptive transfer of antigen-specific T cells. Cancer Immunol Immunother 2010; 59:195-202. [PMID: 19847424 PMCID: PMC11030699 DOI: 10.1007/s00262-009-0778-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 09/24/2009] [Indexed: 10/20/2022]
Abstract
Identification and purification of antigen-specific T cells without altering their functional status are of high scientific and clinical interest. Staining with major histocompatibility complex (MHC)-peptide multimers constitutes a very powerful method to study antigen-specific T-cell subpopulations, allowing their direct visualization and quantification. MHC-peptide multimers, such as dimers, tetramers, pentamers, streptamers, dextramers and octamers have been used to evaluate the frequency of CD8(+) T cells, specific for tumor/leukemia-associated antigens as well as for viral antigens, e.g., CMVpp65 and EBV-EBNA. Moreover, MHC-peptide multimers have been used for rapid and efficient ex vivo isolation and expansion of T cells. A recent development in the field of MHC-peptide multimers led to the purification of CD8(+) T cells specific for leukemia antigens. This might help to select leukemia-specific donor lymphocyte infusions (DLIs), thus allowing dissection of the noxious graft-versus-host disease (GvHD) from beneficial anti-viral and even anti-leukemic effects. This review covers different types of MHC-peptide multimers and their applications, as well as the impact that multimers might have on further development of DLIs.
Collapse
Affiliation(s)
- Rosaely Casalegno-Garduño
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| | - Anita Schmitt
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| | - Junxia Yao
- Center for Stem Cell Research and Application, Institute of Hematology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xinchao Wang
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
- Department of Oncology and Hematology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xun Xu
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Mathias Freund
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| | - Michael Schmitt
- Department of Internal Medicine III, Clinical Stem Cell Transplantation and Immunotherapy, University Clinic Rostock, 18055 Rostock, Germany
| |
Collapse
|
41
|
Wooldridge L, Lissina A, Cole DK, van den Berg HA, Price DA, Sewell AK. Tricks with tetramers: how to get the most from multimeric peptide-MHC. Immunology 2009; 126:147-64. [PMID: 19125886 PMCID: PMC2632693 DOI: 10.1111/j.1365-2567.2008.02848.x] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Revised: 03/18/2008] [Accepted: 03/18/2008] [Indexed: 01/16/2023] Open
Abstract
The development of fluorochrome-conjugated peptide-major histocompatibility complex (pMHC) multimers in conjunction with continuing advances in flow cytometry has transformed the study of antigen-specific T cells by enabling their visualization, enumeration, phenotypic characterization and isolation from ex vivo samples. Here, we bring together and discuss some of the 'tricks' that can be used to get the most out of pMHC multimers. These include: (1) simple procedures that can substantially enhance the staining intensity of cognate T cells with pMHC multimers; (2) the use of pMHC multimers to stain T cells with very-low-affinity T-cell receptor (TCR)/pMHC interactions, such as those that typically predominate in tumour-specific responses; and (3) the physical grading and clonotypic dissection of antigen-specific T cells based on the affinity of their cognate TCR using mutant pMHC multimers in conjunction with new approaches to the molecular analysis of TCR gene expression. We also examine how soluble pMHC can be used to examine T-cell activation, manipulate T-cell responses and study allogeneic and superantigen interactions with TCRs. Finally, we discuss the problems that arise with pMHC class II (pMHCII) multimers because of the low affinity of TCR/pMHCII interactions and lack of 'coreceptor help'.
Collapse
Affiliation(s)
- Linda Wooldridge
- Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Henry Wellcome Building, Heath Park, Cardiff, UK
| | | | | | | | | | | |
Collapse
|
42
|
Chattopadhyay PK, Melenhorst JJ, Ladell K, Gostick E, Scheinberg P, Barrett AJ, Wooldridge L, Roederer M, Sewell AK, Price DA. Techniques to improve the direct ex vivo detection of low frequency antigen-specific CD8+ T cells with peptide-major histocompatibility complex class I tetramers. Cytometry A 2008; 73:1001-9. [PMID: 18836993 DOI: 10.1002/cyto.a.20642] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The ability to quantify and characterize antigen-specific CD8+ T cells irrespective of functional readouts using fluorochrome-conjugated peptide-major histocompatibility complex class I (pMHCI) tetramers in conjunction with flow cytometry has transformed our understanding of cellular immune responses over the past decade. In the case of prevalent CD8+ T cell populations that engage cognate pMHCI tetramers with high avidities, direct ex vivo identification and subsequent data interpretation is relatively straightforward. However, the accurate identification of low frequency antigen-specific CD8+ T cell populations can be complicated, especially in situations where T cell receptor-mediated tetramer binding occurs at low avidities. Here, we highlight a few simple techniques that can be employed to improve the visual resolution, and hence the accurate quantification, of tetramer binding CD8+ T cell populations by flow cytometry. These methodological modifications enhance signal intensity, especially in the case of specific CD8+ T cell populations that bind cognate antigen with low avidities, minimize background noise, and enable improved discrimination of true pMHCI tetramer binding events from nonspecific uptake.
Collapse
Affiliation(s)
- Pratip K Chattopadhyay
- Immunotechnology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
High avidity myeloid leukemia-associated antigen-specific CD8+ T cells preferentially reside in the bone marrow. Blood 2008; 113:2238-44. [PMID: 18997173 DOI: 10.1182/blood-2008-04-151969] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The activity of allogeneic CD8(+) T cells specific for leukemia-associated antigens (LAAs) is thought to mediate, at least in part, the curative effects of hematopoietic stem cell transplantation (HSCT) in myeloid malignancies. However, the identity and nature of clinically relevant LAA-specific CD8(+) T-cell populations have proven difficult to define. Here, we used a combination of coreceptor-mutated peptide-major histocompatibility complex class I (pMHCI) tetramers and polychromatic flow cytometry to examine the avidity profiles, phenotypic characteristics, and anatomical distribution of HLA A*0201-restricted CD8(+) T-cell populations specific for LAAs that are over-expressed in myeloid leukemias. Remarkably, LAA-specific CD8(+) T-cell populations, regardless of fine specificity, were confined almost exclusively to the bone marrow; in contrast, CD8(+) T-cell populations specific for the HLA A*0201-restricted cytomegalovirus (CMV) pp65(495-503) epitope were phenotypically distinct and evenly distributed between bone marrow and peripheral blood. Furthermore, bone marrow-resident LAA-specific CD8(+) T cells frequently engaged cognate antigen with high avidity; notably, this was the case in all tested bone marrow samples derived from patients who achieved clinical remission after HSCT. These data suggest that concomitant examination of bone marrow specimens in patients with myeloid leukemias might yield more definitive information in the search for immunologic prognosticators of clinical outcome.
Collapse
|
44
|
Melenhorst JJ, Scheinberg P, Chattopadhyay PK, Lissina A, Gostick E, Cole DK, Wooldridge L, van den Berg HA, Bornstein E, Hensel NF, Douek DC, Roederer M, Sewell AK, Barrett AJ, Price DA. Detection of low avidity CD8(+) T cell populations with coreceptor-enhanced peptide-major histocompatibility complex class I tetramers. J Immunol Methods 2008; 338:31-9. [PMID: 18675271 PMCID: PMC2714739 DOI: 10.1016/j.jim.2008.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2008] [Revised: 05/08/2008] [Accepted: 07/01/2008] [Indexed: 10/21/2022]
Abstract
The development of soluble recombinant peptide-major histocompatibility complex class I (pMHCI) molecules conjugated in multimeric form to fluorescent labels has enabled the physical quantification and characterization of antigen-specific CD8(+) T cell populations by flow cytometry. Several factors determine the binding threshold that enables visualization of cognate CD8(+) T cells with these reagents; these include the affinity of the T cell receptor (TCR) for pMHCI antigen. Here, we show that multimers constructed from peptide-human leukocyte antigen (pHLA) A0201 monomers engineered in the heavy chain alpha2 domain to enhance CD8 binding (K(D) approximately 85 microM) without impacting the TCR binding platform can detect cognate CD8(+) T cells bearing low affinity TCRs that are not visible with the corresponding wildtype pHLA A0201 multimeric complexes. Mechanistically, this effect is mediated by a disproportionate enhancement of the TCR/pMHCI association rate. In direct ex vivo applications, these coreceptor-enhanced multimers exhibit faithful cognate binding properties; concomitant increases in background staining within the non-cognate CD8(+) T cell population can be resolved phenotypically using polychromatic flow cytometry as a mixture of naïve and memory cells. These findings provide the first validation of a novel approach to the physical detection of low avidity antigen-specific CD8(+) T cell populations; such coreceptor-enhanced multimeric reagents are likely to be useful in a multitude of settings for the detection of auto-immune, tumor-specific and cross-reactive CD8(+) T cells.
Collapse
Affiliation(s)
- J. Joseph Melenhorst
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Phillip Scheinberg
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pratip K. Chattopadhyay
- Immunotechnology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anna Lissina
- Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Emma Gostick
- Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - David K. Cole
- Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Linda Wooldridge
- Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | | | - Ethan Bornstein
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nancy F. Hensel
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mario Roederer
- Immunotechnology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew K. Sewell
- Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - A. John Barrett
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A. Price
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Medical Biochemistry and Immunology, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
45
|
Sartorius R, Pisu P, D'Apice L, Pizzella L, Romano C, Cortese G, Giorgini A, Santoni A, Velotti F, De Berardinis P. The use of filamentous bacteriophage fd to deliver MAGE-A10 or MAGE-A3 HLA-A2-restricted peptides and to induce strong antitumor CTL responses. THE JOURNAL OF IMMUNOLOGY 2008; 180:3719-28. [PMID: 18322177 DOI: 10.4049/jimmunol.180.6.3719] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Delivery of tumor-associated Ag-derived peptides in a high immunogenic form represents one of the key issues for effective peptide-based cancer vaccine development. We report herein the ability of nonpathogenic filamentous bacteriophage fd virions to deliver HLA-A2-restricted MAGE-A10(254-262)- or MAGE-A3(271-279)-derived peptides and to elicit potent specific CTL responses in vitro and in vivo. Interestingly, human anti-MAGE-A3(271-279)-specific CTLs were able to kill human MAGE-A3(+) tumor cells, even if these cells naturally express a low amount of MAGE-A3(271-279) peptide-HLA epitope surface complexes and are usually not recognized by CTLs generated by conventional stimulation procedures. MAGE-A3(271-279)-specific/CD8(+) CTL clones were isolated from in vitro cultures, and their high avidity for Ag recognition was assessed. Moreover, in vivo tumor protection assay showed that vaccination of humanized HHD (HLA-A2.1(+)/H2-D(b+)) transgenic mice with phage particles expressing MAGE-A3(271-279)-derived peptides hampered tumor growth. Overall, these data indicate that engineered filamentous bacteriophage virions increase substantially the immunogenicity of delivered tumor-associated Ag-derived peptides, thus representing a novel powerful system for the development of effective peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Rossella Sartorius
- Institute of Protein Biochemistry, Consiglio Nazionale delle Ricerche, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
A T-cell receptor associated with naturally occurring human tumor immunity. Proc Natl Acad Sci U S A 2008; 104:19073-8. [PMID: 18045792 DOI: 10.1073/pnas.0704336104] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The onconeural antigens appear to serve as tumor rejection antigens in the paraneoplastic neurologic disorders. Here, we used an unbiased peptide binding screen, followed by studies in HLA-A2.1 transgenic mice to identify naturally processed HLA-A2.1 restricted epitopes of the paraneoplastic cerebellar degeneration breast/ovarian cancer antigen cdr2. These mice were used to clone high-avidity cdr2-specific CD8(+) T cells that recognize human tumor cells presenting endogenously loaded MHC class I-cdr2 peptide. T cells with this specificity were detected in the peripheral blood of two HLA-A2.1(+) paraneoplastic cerebellar degeneration patients. We cloned T cell receptor (TCR) alpha and beta genes from cdr2-specific T cells; electroporation of RNA encoding this TCR turned nonreactive donor T cells into efficient killers of human cdr2-expressing tumor cells. Cloned cdr2-specific TCR genes provide a clinically relevant means for immunologic targeting of human gynecologic cancers.
Collapse
|
47
|
High pro-inflammatory cytokine secretion and loss of high avidity cross-reactive cytotoxic T-cells during the course of secondary dengue virus infection. PLoS One 2007; 2:e1192. [PMID: 18060049 PMCID: PMC2092391 DOI: 10.1371/journal.pone.0001192] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Accepted: 10/19/2007] [Indexed: 11/23/2022] Open
Abstract
Background Dengue is one of the most important human diseases transmitted by an arthropod vector and the incidence of dengue virus infection has been increasing – over half the world's population now live in areas at risk of infection. Most infections are asymptomatic, but a subset of patients experience a potentially fatal shock syndrome characterised by plasma leakage. Severe forms of dengue are epidemiologically associated with repeated infection by more than one of the four dengue virus serotypes. Generally attributed to the phenomenon of antibody-dependent enhancement, recent observations indicate that T-cells may also influence disease phenotype. Methods and Findings Virus-specific cytotoxic T lymphocytes (CTL) showing high level cross reactivity between dengue serotypes could be expanded from blood samples taken during the acute phase of secondary dengue infection. These could not be detected in convalescence when only CTL populations demonstrating significant serotype specificity were identified. Dengue cross-reactive CTL clones derived from these patients were of higher avidity than serotype-specific clones and produced much higher levels of both type 1 and certain type 2 cytokines, many previously implicated in dengue pathogenesis. Conclusion Dengue serotype cross-reactive CTL clones showing high avidity for antigen produce higher levels of inflammatory cytokines than serotype-specific clones. That such cells cannot be expanded from convalescent samples suggests that they may be depleted, perhaps as a consequence of activation-induced cell death. Such high avidity cross-reactive memory CTL may produce inflammatory cytokines during the course of secondary infection, contributing to the pathogenesis of vascular leak. These cells appear to be subsequently deleted leaving a more serotype-specific memory CTL pool. Further studies are needed to relate these cellular observations to disease phenotype in a large group of patients. If confirmed they have significant implications for understanding the role of virus-specific CTL in pathogenesis of dengue disease.
Collapse
|
48
|
Iero M, Squarcina P, Romero P, Guillaume P, Scarselli E, Cerino R, Carrabba M, Toutirais O, Parmiani G, Rivoltini L. Low TCR avidity and lack of tumor cell recognition in CD8(+) T cells primed with the CEA-analogue CAP1-6D peptide. Cancer Immunol Immunother 2007; 56:1979-91. [PMID: 17564703 PMCID: PMC11030693 DOI: 10.1007/s00262-007-0342-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Accepted: 05/17/2007] [Indexed: 12/22/2022]
Abstract
The use of "altered peptide ligands" (APL), epitopes designed for exerting increased immunogenicity as compared with native determinants, represents nowadays one of the most utilized strategies for overcoming immune tolerance to self-antigens and boosting anti-tumor T cell-mediated immune responses. However, the actual ability of APL-primed T cells to cross-recognize natural epitopes expressed by tumor cells remains a crucial concern. In the present study, we show that CAP1-6D, a superagonist analogue of a carcinoembriyonic antigen (CEA)-derived HLA-A*0201-restricted epitope widely used in clinical setting, reproducibly promotes the generation of low-affinity CD8(+) T cells lacking the ability to recognized CEA-expressing colorectal carcinoma (CRC) cells. Short-term T cell cultures, obtained by priming peripheral blood mononuclear cells from HLA-A*0201(+) healthy donors or CRC patients with CAP1-6D, were indeed found to heterogeneously cross-react with saturating concentrations of the native peptide CAP1, but to fail constantly lysing or recognizing through IFN- gamma release CEA(+)CRC cells. Characterization of anti-CAP1-6D T cell avidity, gained through peptide titration, CD8-dependency assay, and staining with mutated tetramers (D227K/T228A), revealed that anti-CAP1-6D T cells exerted a differential interaction with the two CEA epitopes, i.e., displaying high affinity/CD8-independency toward the APL and low affinity/CD8-dependency toward the native CAP1 peptide. Our data demonstrate that the efficient detection of self-antigen expressed by tumors could be a feature of high avidity CD8-independent T cells, and underline the need for extensive analysis of tumor cross-recognition prior to any clinical usage of APL as anti-cancer vaccines.
Collapse
Affiliation(s)
- Manuela Iero
- Unit of Immunotherapy of Human Tumors, IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Paola Squarcina
- Unit of Immunotherapy of Human Tumors, IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Pedro Romero
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Philippe Guillaume
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Elisa Scarselli
- Molecular and Cellular Biology Department, IRBM P. Angeletti, Pomezia, Italy
| | - Raffaele Cerino
- Molecular and Cellular Biology Department, IRBM P. Angeletti, Pomezia, Italy
| | - Matteo Carrabba
- Unit of Immunotherapy of Human Tumors, IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Olivier Toutirais
- Laboratoire de Cytogénétique et de Biologie Cellulaire, Hôpital Pontchaillou, Rennes, France
| | - Giorgio Parmiani
- Unit of Immunotherapy of Human Tumors, IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
- Present Address: Unit of Immunobiotherapy of Solid Tumors, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| |
Collapse
|
49
|
Immunization with a lentivector that targets tumor antigen expression to dendritic cells induces potent CD8+ and CD4+ T-cell responses. J Virol 2007; 82:86-95. [PMID: 17959670 DOI: 10.1128/jvi.01289-07] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lentivectors stimulate potent immune responses to antigen transgenes and are being developed as novel genetic vaccines. To improve safety while retaining efficacy, we constructed a lentivector in which transgene expression was restricted to antigen-presenting cells using the mouse dectin-2 gene promoter. This lentivector expressed a green fluorescent protein (GFP) transgene in mouse bone marrow-derived dendritic cell cultures and in human skin-derived Langerhans and dermal dendritic cells. In mice GFP expression was detected in splenic dectin-2(+) cells after intravenous injection and in CD11c(+) dendritic cells in the draining lymph node after subcutaneous injection. A dectin-2 lentivector encoding the human melanoma antigen NY-ESO-1 primed an NY-ESO-1-specific CD8(+) T-cell response in HLA-A2 transgenic mice and stimulated a CD4(+) T-cell response to a newly identified NY-ESO-1 epitope presented by H2 I-A(b). As immunization with the optimal dose of the dectin-2 lentivector was similar to that stimulated by a lentivector containing a strong constitutive viral promoter, targeting antigen expression to dendritic cells can provide a safe and effective vaccine.
Collapse
|
50
|
Schaubert KL, Price DA, Frahm N, Li J, Ng HL, Joseph A, Paul E, Majumder B, Ayyavoo V, Gostick E, Adams S, Marincola FM, Sewell AK, Altfeld M, Brenchley JM, Douek DC, Yang OO, Brander C, Goldstein H, Kan-Mitchell J. Availability of a diversely avid CD8+ T cell repertoire specific for the subdominant HLA-A2-restricted HIV-1 Gag p2419-27 epitope. THE JOURNAL OF IMMUNOLOGY 2007; 178:7756-66. [PMID: 17548613 PMCID: PMC2365726 DOI: 10.4049/jimmunol.178.12.7756] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HLA-A2-restricted CTL responses to immunodominant HIV-1 epitopes do not appear to be very effective in the control of viral replication in vivo. In this study, we studied human CD8+ T cell responses to the subdominant HLA-A2-restricted epitope TV9 (Gag p24(19-27), TLNAWVKVV) to explore the possibility of increasing its immune recognition. We confirmed in a cohort of 313 patients, infected by clade B or clade C viruses, that TV9 is rarely recognized. Of interest, the functional sensitivity of the TV9 response can be relatively high. The potential T cell repertoires for TV9 and the characteristics of constituent clonotypes were assessed by ex vivo priming of circulating CD8+ T cells from healthy seronegative donors. TV9-specific CTLs capable of suppressing viral replication in vitro were readily generated, suggesting that the cognate T cell repertoire is not limiting. However, these cultures contained multiple discrete populations with a range of binding avidities for the TV9 tetramer and correspondingly distinct functional dependencies on the CD8 coreceptor. The lack of dominant clonotypes was not affected by the stage of maturation of the priming dendritic cells. Cultures primed by dendritic cells transduced to present endogenous TV9 were also incapable of clonal maturation. Thus, a diffuse TCR repertoire appeared to be an intrinsic characteristic of TV9-specific responses. These data indicate that subdominance is not a function of poor immunogenicity, cognate TCR repertoire availability, or the potential avidity properties thereof, but rather suggest that useful responses to this epitope are suppressed by competing CD8+ T cell populations during HIV-1 infection.
Collapse
Affiliation(s)
- Keri L. Schaubert
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Biological Sciences, University of Texas, El Paso, TX 79968
| | - David A. Price
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- Weatherall Institute of Molecular Medicine, University of Oxford, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Nicole Frahm
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Charles-town, MA 02192
| | - Jinzhu Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Hwee L. Ng
- Department of Medicine and AIDS Institute, Center for Health Sciences, University of California, Los Angeles, CA 90095
| | - Aviva Joseph
- Department of Micro-biology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Elyse Paul
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Biswanath Majumder
- Department of Infectious Diseases and Micro-biology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261
| | - Velpandi Ayyavoo
- Department of Infectious Diseases and Micro-biology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261
| | - Emma Gostick
- Weatherall Institute of Molecular Medicine, University of Oxford, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Sharon Adams
- Section of Immunogenetics, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD 20892
| | - Francesco M. Marincola
- Section of Immunogenetics, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD 20892
| | - Andrew K. Sewell
- Department of Medical Biochemistry and Immunology, Cardiff University, Cardiff, Wales, United Kingdom
| | - Marcus Altfeld
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Charles-town, MA 02192
| | - Jason M. Brenchley
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Otto O. Yang
- Department of Medicine and AIDS Institute, Center for Health Sciences, University of California, Los Angeles, CA 90095
| | - Christian Brander
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Charles-town, MA 02192
| | - Harris Goldstein
- Department of Micro-biology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - June Kan-Mitchell
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Biological Sciences, University of Texas, El Paso, TX 79968
- Address correspondence and reprint requests to Dr. June Kan-Mitchell, Biological Sciences Building, University of Texas, 500 West University Avenue, El Paso, TX 79968. E-mail address:
| |
Collapse
|