1
|
Wellach K, Riemer AB. Highly sensitive live-cell imaging-based cytotoxicity assay enables functional validation of rare epitope-specific CTLs. Front Immunol 2025; 16:1558620. [PMID: 40406125 PMCID: PMC12095279 DOI: 10.3389/fimmu.2025.1558620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Many immunotherapeutic approaches aim to induce epitope-specific T-cell cytotoxicity. However, the identification-and especially the functional validation-of suitable epitopes by in vitro cytotoxicity assays can be challenging, particularly when the number of available epitope-specific cytotoxic T cells (CTLs) is limited. Here, we present a highly sensitive image-based cytotoxicity assay that allows the functional analysis of rare epitope-specific T cells. The live-cell imaging-based setup combines transient red labeling of target cells with a green caspase 3/7 probe, allowing reliable measurement of the fraction of apoptotic target cells. Time-course analysis enables the monitoring of subtle differences. This highly flexible assay can be applied to assess the killing of either target cells with endogenous epitope presentation or those artificially loaded with the epitope of interest. Analysis of assay sensitivity demonstrated that cytotoxicity mediated by as few as 0.1% epitope-specific CTLs in a T-cell culture can still be detected. The epitope-specificity of the assay was additionally validated by specific upregulation of PD-1 and LAG-3 on epitope-specific T cells, as well as the epitope-specific induction of interferon-γ release. Finally, the assay was successfully applied to functionally validate human papillomavirus (HPV)16 epitopes, by detecting epitope-specific killing of established patient-derived tumor cell lines by rare T-cell populations expanded from peripheral blood. Overall, this cytotoxicity assay setup provides a straightforward approach to assess the cytotoxic capacity of rare epitope-specific T cells and enables the analysis of T-cell responses against endogenously presented epitopes.
Collapse
Affiliation(s)
- Kathrin Wellach
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Center for Infection Research (DZIF), Molecular Vaccine Design, Partner Site Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Angelika B. Riemer
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Center for Infection Research (DZIF), Molecular Vaccine Design, Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
2
|
Nguyen KA, Liu Z, Davies JS, McIntosh CP, Draper LM, Norberg SM, Rae Z, Achar SR, Altan-Bonnet G, Zhang L, Wu X, Meyer TJ, Kelly MC, Taylor N, Hinrichs CS, Ishii K. CD22 TCR-engineered T cells exert antileukemia cytotoxicity without causing inflammatory responses. SCIENCE ADVANCES 2025; 11:eadq4297. [PMID: 40203088 PMCID: PMC11980841 DOI: 10.1126/sciadv.adq4297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Chimeric antigen receptor (CAR) T cells effectively treat B cell malignancies. However, CAR-T cells cause inflammatory toxicities such as cytokine release syndrome (CRS), which is in contrast to T cell receptor (TCR)-engineered T cells against various antigens that historically have rarely been associated with CRS. To study whether and how differences in receptor types affect the propensity for eliciting inflammatory responses in a model system wherein TCR and CAR target equalized sources of clinically relevant antigen, we discovered a CD22-specific TCR and compared it to CD22 CAR. Both CD22 TCR-T and CD22 CAR-T cells eradicated leukemia in xenografts, but only CD22 CAR-T cells induced dose-dependent systemic inflammation. Compared to TCR-T cells, CAR-T cells disproportionately upregulated inflammatory pathways without concordant augmentation in pathways involved in direct cytotoxicity upon antigen engagement. These differences in antileukemia responses comparing TCR-T and CAR-T cells highlight the potential opportunity to improve therapeutic safety by using TCRs.
Collapse
MESH Headings
- Humans
- Animals
- Sialic Acid Binding Ig-like Lectin 2/immunology
- Sialic Acid Binding Ig-like Lectin 2/genetics
- Sialic Acid Binding Ig-like Lectin 2/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Mice
- Inflammation/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Leukemia/therapy
- Leukemia/immunology
- Leukemia/pathology
- Cytotoxicity, Immunologic
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kilyna A. Nguyen
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zhihui Liu
- Pediatric Oncology Branch, CCR, NCI, NIH, Bethesda, MD, USA
| | - John S. Davies
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA, USA
| | - Crystal P. McIntosh
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Lindsey M. Draper
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Scott M. Norberg
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zachary Rae
- Single Cell Analysis Facility, CCR, NCI, NIH, Bethesda, MD, USA
| | - Sooraj R. Achar
- Laboratory of Integrative Cancer Immunology, CCR, NCI, NIH, Bethesda, MD, USA
| | | | - Ling Zhang
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Xiaolin Wu
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, NCI, NIH, Frederick, MD, USA
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, CCR, NCI, NIH, Bethesda, MD, USA
| | | | - Naomi Taylor
- Pediatric Oncology Branch, CCR, NCI, NIH, Bethesda, MD, USA
| | - Christian S. Hinrichs
- Duncan and Nancy MacMillan Center of Excellence in Cancer Immunotherapy and Metabolism, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Kazusa Ishii
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
3
|
Krämer C, Kilian M, Chih YC, Kourtesakis A, Hoffmann DC, Boschert T, Koopmann P, Sanghvi K, De Roia A, Jung S, Jähne K, Day B, Shultz LD, Ratliff M, Harbottle R, Green EW, Will R, Wick W, Platten M, Bunse L. NLGN4X TCR transgenic T cells to treat gliomas. Neuro Oncol 2024; 26:266-278. [PMID: 37715782 PMCID: PMC10836769 DOI: 10.1093/neuonc/noad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Neuroligin 4 X-linked (NLGN4X) harbors a human leukocyte antigen (HLA)-A*02-restricted tumor-associated antigen, overexpressed in human gliomas, that was found to induce specific cytotoxic T cell responses following multi-peptide vaccination in patients with newly diagnosed glioblastoma. METHODS T cell receptor (TCR) discovery was performed using droplet-based single-cell TCR sequencing of NLGN4X-tetramer-sorted T cells postvaccination. The identified TCR was delivered to Jurkat T cells and primary human T cells (NLGN4X-TCR-T). Functional profiling of NLGN4X-TCR-T was performed by flow cytometry and cytotoxicity assays. Therapeutic efficacy of intracerebroventricular NLGN4X-TCR-T was assessed in NOD scid gamma (NSG) major histocompatibility complex (MHC) I/II knockout (KO) (NSG MHC I/II KO) mice bearing NLGN4X-expressing experimental gliomas. RESULTS An HLA-A*02-restricted vaccine-induced T cell receptor specifically binding NLGN4X131-139 was applied for preclinical therapeutic use. Reactivity, cytotoxicity, and polyfunctionality of this NLGN4X-specific TCR are demonstrated in various cellular models. Intracerebroventricular administration of NLGN4X-TCR-T prolongs survival and leads to an objective response rate of 44.4% in experimental glioma-bearing NSG MHC I/II KO mice compared to 0.0% in control groups. CONCLUSION NLGN4X-TCR-T demonstrate efficacy in a preclinical glioblastoma model. On a global scale, we provide the first evidence for the therapeutic retrieval of vaccine-induced human TCRs for the off-the-shelf treatment of glioblastoma patients.Keywords cell therapy | glioblastoma | T cell receptor | tumor antigen.
Collapse
Affiliation(s)
- Christoper Krämer
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Kilian
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yu-Chan Chih
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
| | - Alexandros Kourtesakis
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
- Neurology Clinic, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
- DKTK CCU Neurooncology, DKFZ, Heidelberg, Germany
| | - Dirk C Hoffmann
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
- Neurology Clinic, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
- DKTK CCU Neurooncology, DKFZ, Heidelberg, Germany
| | - Tamara Boschert
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
- Helmholtz Institute of Translational Oncology (HI-TRON), Mainz, Germany
| | - Philipp Koopmann
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Khwab Sanghvi
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
| | - Alice De Roia
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
- DNA Vector Laboratory, DKFZ, Heidelberg, Germany
| | - Stefanie Jung
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kristine Jähne
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bryan Day
- Faculty of Medicine, University of Queensland, Herston, Australia
- Cell and Molecular Biology Department, Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer MRI, Brisbane, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Gardens Point, Australia
| | - Lenny D Shultz
- Department of Immunology, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Miriam Ratliff
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | | | - Edward W Green
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Will
- Neurology Clinic, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
- DKTK CCU Neurooncology, DKFZ, Heidelberg, Germany
- Core Facility Cellular tools, DKFZ, Heidelberg, Germany
| | | | - Michael Platten
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Helmholtz Institute of Translational Oncology (HI-TRON), Mainz, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Lukas Bunse
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| |
Collapse
|
4
|
Schifflers C, Zottnick S, Förster JD, Kruse S, Yang R, Wiethoff H, Bozza M, Hoppe-Seyler K, Heikenwälder M, Harbottle RP, Michiels C, Riemer AB. Development of an Orthotopic HPV16-Dependent Base of Tongue Tumor Model in MHC-Humanized Mice. Pathogens 2023; 12:pathogens12020188. [PMID: 36839460 PMCID: PMC9958775 DOI: 10.3390/pathogens12020188] [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: 11/17/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) caused by infections with high-risk human papillomaviruses (HPV) are responsible for an increasing number of head and neck cancers, particularly in the oropharynx. Despite the significant biological differences between HPV-driven and HPV-negative HNSCC, treatment strategies are similar and not HPV targeted. HPV-driven HNSCC are known to be more sensitive to treatment, particularly to radiotherapy, which is at least partially due to HPV-induced immunogenicity. The development of novel therapeutic strategies that are specific for HPV-driven cancers requires tumor models that reflect as closely as possible the characteristics and complexity of human tumors and their response to treatment. Current HPV-positive cancer models lack one or more hallmarks of their human counterpart. This study presents the development of a new HPV16 oncoprotein-dependent tumor model in MHC-humanized mice, modeling the major biologic features of HPV-driven tumors and presenting HLA-A2-restricted HPV16 epitopes. Furthermore, this model was developed to be orthotopic (base of tongue). Thus, it also reflects the correct tumor microenvironment of HPV-driven HNSCC. The cancer cells are implanted in a manner that allows the exact control of the anatomical location of the developing tumor, thereby homogenizing tumor growth. In conclusion, the new model is suited to study HPV16-specific therapeutic vaccinations and other immunotherapies, as well as tumor-targeted interventions, such as surgery or radiotherapy, or a combination of all these modalities.
Collapse
Affiliation(s)
- Christoph Schifflers
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Cell Biology Research Unit (URBC)–Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Samantha Zottnick
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research, Partner Site Heidelberg, 69120 Heidelberg, Germany
| | - Jonas D. Förster
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research, Partner Site Heidelberg, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Sebastian Kruse
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research, Partner Site Heidelberg, 69120 Heidelberg, Germany
| | - Ruwen Yang
- Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hendrik Wiethoff
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Helmholtz-University Group Cell Plasticity and Epigenetic Remodeling, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Matthias Bozza
- DNA Vector Laboratory, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Karin Hoppe-Seyler
- Molecular Therapy of Virus-Associated Cancers, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mathias Heikenwälder
- Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Richard P. Harbottle
- DNA Vector Laboratory, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Carine Michiels
- Cell Biology Research Unit (URBC)–Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Angelika B. Riemer
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research, Partner Site Heidelberg, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-42-3820
| |
Collapse
|
5
|
Mohan N, Wellach K, Özerdem C, Veits N, Förster JD, Foehr S, Bonsack M, Riemer AB. Effects of hypoxia on antigen presentation and T cell-based immune recognition of HPV16-transformed cells. Front Immunol 2022; 13:918528. [PMID: 36341354 PMCID: PMC9634485 DOI: 10.3389/fimmu.2022.918528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/26/2022] [Indexed: 02/27/2025] Open
Abstract
Attempts to develop a therapeutic vaccine against human papillomavirus (HPV)-induced malignancies have mostly not been clinically successful to date. One reason may be the hypoxic microenvironment present in most tumors, including cervical cancer. Hypoxia dysregulates the levels of human leukocyte antigen (HLA) class I molecules in different tumor entities, impacts the function of cytotoxic T cells, and leads to decreased protein levels of the oncoproteins E6 and E7 in HPV-transformed cells. Therefore, we investigated the effect of hypoxia on the presentation of HPV16 E6- and E7-derived epitopes in cervical cancer cells and its effect on epitope-specific T cell cytotoxicity. Hypoxia induced downregulation of E7 protein levels in all analyzed cell lines, as assessed by Western blotting. However, contrary to previous reports, no perturbation of antigen processing and presentation machinery (APM) components and HLA-A2 surface expression upon hypoxia treatment was detected by mass spectrometry and flow cytometry, respectively. Cytotoxicity assays performed in hypoxic conditions showed differential effects on the specific killing of HPV16-positive cervical cancer cells by epitope-specific CD8+ T cell lines in a donor- and peptide-specific manner. Effects of hypoxia on the expression of PD-L1 were ruled out by flow cytometry analysis. Altogether, our results under hypoxia show a decreased expression of E6 and E7, but an intact APM, and epitope- and donor-dependent effects on T cell cytotoxicity towards HPV16-positive target cells. This suggests that successful immunotherapies can be developed for hypoxic HPV-induced cervical cancer, with careful choice of target epitopes, and ideally in combination with hypoxia-alleviating measures.
Collapse
Affiliation(s)
- Nitya Mohan
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Kathrin Wellach
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Ceren Özerdem
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Nisha Veits
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jonas D. Förster
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Sophia Foehr
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Maria Bonsack
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Angelika B. Riemer
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
6
|
Olivo Pimentel V, Yaromina A, Marcus D, Dubois LJ, Lambin P. A novel co-culture assay to assess anti-tumor CD8 + T cell cytotoxicity via luminescence and multicolor flow cytometry. J Immunol Methods 2020; 487:112899. [PMID: 33068606 DOI: 10.1016/j.jim.2020.112899] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/16/2020] [Accepted: 10/11/2020] [Indexed: 12/31/2022]
Abstract
T cell immunotherapies have shown great promise in patients with advanced cancer disease, revolutionizing treatment. T cell cytotoxicity is crucial in its efficacy, therefore developing ex vivo methods testing tumor and T cell interactions is pivotal. Increasing efforts have been made in developing co-culture assays with sophisticated materials and platforms aiming to mimic the tumor microenvironment (TME), but its complexity makes it difficult to develop the ideal model. In this study, we developed a simple co-culture assay, reproducible in any lab, but respecting the multicellular nature of the TME. Our goal is to combine in a single assay well-established techniques such as a luciferase assay for target cell viability analysis, a CD107a degranulation assay, and multicolor flow cytometry for the detection of cytokines and cytotoxicity markers. Cell suspensions of whole spleens and tumors containing splenic or tumor-infiltrating effector T cells of mice bearing Lewis lung carcinoma (LLC) or CT26 colon carcinoma tumors treated with radiation alone or in combination with immunotherapies were used for co-culture. LLC and CT26 cell lines transduced with the firefly luciferase gene were used as target cells. We demonstrated that splenocytes and tumor-infiltrating T cells derived from mice treated with combination therapy were able to kill approximately 50% of target cells after 48 h of co-culture. This effect was tumor cell-specific and dependent on CD8+ T cells evidenced by in vitro CD8+ T cell depletion. Flow cytometry demonstrated increased expression of CD107a and production of granzyme B, IFNγ, and TNFα by CD8+ T cells. Our co-culture assay is therefore suitable as proof of principle for in vivo therapeutic studies testing immunotherapies, and specifically to assess the involvement of cytotoxic CD8+ T cells in treatment response in LLC and CT26 tumor models. We also propose this assay as an ex vivo platform for high-throughput screening of immunomodulating agents to be tested in these two murine tumor models. This assay can be adapted to other tumor models after optimizations.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/therapy
- Cell Line, Tumor
- Coculture Techniques
- Colonic Neoplasms/immunology
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Cytotoxicity, Immunologic
- Flow Cytometry
- Granzymes/metabolism
- Immunotherapy
- Interferon-gamma/metabolism
- Luciferases, Firefly/biosynthesis
- Luciferases, Firefly/genetics
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lysosomal Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Proof of Concept Study
- Radiotherapy
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Tumor Microenvironment
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Verónica Olivo Pimentel
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Damiënne Marcus
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
7
|
Teck AT, Urban S, Quass P, Nelde A, Schuster H, Letsch A, Busse A, Walz JS, Keilholz U, Ochsenreither S. Cancer testis antigen Cyclin A1 harbors several HLA-A*02:01-restricted T cell epitopes, which are presented and recognized in vivo. Cancer Immunol Immunother 2020; 69:1217-1227. [PMID: 32157447 PMCID: PMC8222032 DOI: 10.1007/s00262-020-02519-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/12/2020] [Indexed: 01/22/2023]
Abstract
Cyclin A1 is a promising antigen for T cell therapy being selectively expressed in high-grade ovarian cancer (OC) and acute myeloid leukemia (AML) stem cells. For adoptive T cell therapy, a single epitope has to be selected, with high affinity to MHC class I and adequate processing and presentation by malignant cells to trigger full activation of specific T cells. In silico prediction with three algorithms indicated 13 peptides of Cyclin A1 9 to 11 amino acids of length to have high affinity to HLA-A*02:01. Ten of them proved to be affine in an HLA stabilization assay using TAP-deficient T2 cells. Their immunogenicity was assessed by repetitive stimulation of CD8+ T cells from two healthy donors with single-peptide-pulsed dendritic cells or monocytes. Intracellular cytokine staining quantified the enrichment of peptide-specific functional T cells. Seven peptides were immunogenic, three of them against both donors. Specific cell lines were cloned and used in killing assays to demonstrate recognition of endogenous Cyclin A1 in the HLA-A*02:01-positive AML cell line THP-1. Immunopeptidome analysis based on direct isolation of HLA-presented peptides by mass spectrometry of primary AML and OC samples identified four naturally presented epitopes of Cyclin A1. The immunopeptidome of HeLa cells transfected with Cyclin A1 and HLA-A*02:01 revealed six Cyclin A1-derived HLA ligands. Epitope p410–420 showed high affinity to HLA-A*02:01 and immunogenicity in both donors. It proved to be naturally presented on primary AML blast and provoked spontaneous functional response of T cells from treatment naïve OC and, therefore, warrants further development for clinical application.
Collapse
Affiliation(s)
- Anja Tatjana Teck
- Department of Hematology and Oncology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany
| | - Sabrina Urban
- Department of Hematology and Oncology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany
| | - Petra Quass
- Department of Hematology and Oncology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany.,Charité Comprehensive Cancer Center, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annika Nelde
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.,Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tübingen, Tübingen, Germany
| | - Heiko Schuster
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.,Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Anne Letsch
- Department of Hematology and Oncology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antonia Busse
- Department of Hematology and Oncology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juliane Sarah Walz
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Ochsenreither
- Department of Hematology and Oncology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany. .,Charité Comprehensive Cancer Center, Charitéplatz 1, 10117, Berlin, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
8
|
Rapid Assessment of Functional Avidity of Tumor-Specific T Cell Receptors Using an Antigen-Presenting Tumor Cell Line Electroporated with Full-Length Tumor Antigen mRNA. Cancers (Basel) 2020; 12:cancers12020256. [PMID: 31972992 PMCID: PMC7072428 DOI: 10.3390/cancers12020256] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/22/2022] Open
Abstract
The functional avidity of T-cell receptor (TCR)-engineered T cells towards their cognate epitope plays a crucial role in successfully targeting and killing tumor cells expressing the tumor-associated antigen (TAA). When evaluating in vitro functional T-cell avidity, an important aspect that is often neglected is the antigen-presenting cell (APC) used in the assay. Cell-based models for antigen-presentation, such as tumor cell lines, represent a valid alternative to autologous APCs due to their availability, off-the-shelf capabilities, and the broad range of possibilities for modification via DNA or messenger RNA (mRNA) transfection. To find a valuable model APC for in vitro validation of TAA Wilms’ tumor 1 (WT1)-specific TCRs, we tested four different WT1 peptide-pulsed HLA-A2+ tumor cell lines commonly used in T-cell stimulation assays. We found the multiple myeloma cell line U266 to be a suitable model APC to evaluate differences in mean functional avidity (EC50) values of transgenic TCRs following transfection in 2D3 Jurkat T cells. Next, to assess the dose-dependent antigen-specific responsiveness of WT1 TCR-engineered 2D3 T cells to endogenously processed epitopes, we electroporated U266 cells with different amounts of full-length antigen WT1 mRNA. Finally, we analyzed the functional avidity of WT1 TCR-transfected primary CD8 T cells towards WT1 mRNA-electroporated U266 cells. In this study, we demonstrate that both the APC and the antigen loading method (peptide pulsing versus full-length mRNA transfection) to analyze T-cell functional avidity have a significant impact on the EC50 values of a given TCR. For rapid assessment of the functional avidity of a cloned TCR towards its endogenously processed MHC I-restricted epitope, we showcase that the TAA mRNA-transfected U266 cell line is a suitable and versatile model APC.
Collapse
|
9
|
Hilf N, Kuttruff-Coqui S, Frenzel K, Bukur V, Stevanović S, Gouttefangeas C, Platten M, Tabatabai G, Dutoit V, van der Burg SH, Thor Straten P, Martínez-Ricarte F, Ponsati B, Okada H, Lassen U, Admon A, Ottensmeier CH, Ulges A, Kreiter S, von Deimling A, Skardelly M, Migliorini D, Kroep JR, Idorn M, Rodon J, Piró J, Poulsen HS, Shraibman B, McCann K, Mendrzyk R, Löwer M, Stieglbauer M, Britten CM, Capper D, Welters MJP, Sahuquillo J, Kiesel K, Derhovanessian E, Rusch E, Bunse L, Song C, Heesch S, Wagner C, Kemmer-Brück A, Ludwig J, Castle JC, Schoor O, Tadmor AD, Green E, Fritsche J, Meyer M, Pawlowski N, Dorner S, Hoffgaard F, Rössler B, Maurer D, Weinschenk T, Reinhardt C, Huber C, Rammensee HG, Singh-Jasuja H, Sahin U, Dietrich PY, Wick W. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 2019; 565:240-245. [PMID: 30568303 DOI: 10.1038/s41586-018-0810-y] [Citation(s) in RCA: 632] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022]
Abstract
Patients with glioblastoma currently do not sufficiently benefit from recent breakthroughs in cancer treatment that use checkpoint inhibitors1,2. For treatments using checkpoint inhibitors to be successful, a high mutational load and responses to neoepitopes are thought to be essential3. There is limited intratumoural infiltration of immune cells4 in glioblastoma and these tumours contain only 30-50 non-synonymous mutations5. Exploitation of the full repertoire of tumour antigens-that is, both unmutated antigens and neoepitopes-may offer more effective immunotherapies, especially for tumours with a low mutational load. Here, in the phase I trial GAPVAC-101 of the Glioma Actively Personalized Vaccine Consortium (GAPVAC), we integrated highly individualized vaccinations with both types of tumour antigens into standard care to optimally exploit the limited target space for patients with newly diagnosed glioblastoma. Fifteen patients with glioblastomas positive for human leukocyte antigen (HLA)-A*02:01 or HLA-A*24:02 were treated with a vaccine (APVAC1) derived from a premanufactured library of unmutated antigens followed by treatment with APVAC2, which preferentially targeted neoepitopes. Personalization was based on mutations and analyses of the transcriptomes and immunopeptidomes of the individual tumours. The GAPVAC approach was feasible and vaccines that had poly-ICLC (polyriboinosinic-polyribocytidylic acid-poly-L-lysine carboxymethylcellulose) and granulocyte-macrophage colony-stimulating factor as adjuvants displayed favourable safety and strong immunogenicity. Unmutated APVAC1 antigens elicited sustained responses of central memory CD8+ T cells. APVAC2 induced predominantly CD4+ T cell responses of T helper 1 type against predicted neoepitopes.
Collapse
Affiliation(s)
- Norbert Hilf
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | - Stefan Stevanović
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
| | - Cécile Gouttefangeas
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Michael Platten
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
- Medical Faculty Mannheim, Mannheim, Germany
| | - Ghazaleh Tabatabai
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
- University Hospital Tübingen, Tübingen, Germany
| | | | - Sjoerd H van der Burg
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Leiden University Medical Center, Leiden, The Netherlands
| | - Per Thor Straten
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Hideho Okada
- University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Arie Admon
- Technion - Israel Institute of Technology, Haifa, Israel
| | | | | | - Sebastian Kreiter
- BioNTech AG, Mainz, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Andreas von Deimling
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | | | | | - Judith R Kroep
- Leiden University Medical Center, Leiden, The Netherlands
| | - Manja Idorn
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jordi Rodon
- Vall d'Hebron University Hospital, Barcelona, Spain
- M. D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | | | | | | | | | | | | | - Monika Stieglbauer
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Cedrik M Britten
- BioNTech AG, Mainz, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Oncology R&D, GlaxoSmithKline, Stevenage, UK
| | - David Capper
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
- Charité, University Medicine Berlin, Berlin, Germany
| | - Marij J P Welters
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Elisa Rusch
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- CIMT/CIP - Association for Cancer Immunotherapy, working group Cancer Immunoguiding Program, Mainz, Germany
| | - Lukas Bunse
- University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Colette Song
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | - Jörg Ludwig
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | - John C Castle
- BioNTech AG, Mainz, Germany
- Agenus Inc., Lexington, KY, USA
| | | | - Arbel D Tadmor
- TRON GmbH - Translational Oncology at the University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Edward Green
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
- Medical Faculty Mannheim, Mannheim, Germany
| | | | - Miriam Meyer
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | - Sonja Dorner
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | | | | | | | - Hans-Georg Rammensee
- Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Partner Site Tübingen, Tübingen, Germany
| | | | | | | | - Wolfgang Wick
- University Hospital Heidelberg, Heidelberg, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
10
|
Kruse S, Büchler M, Uhl P, Sauter M, Scherer P, Lan TCT, Zottnick S, Klevenz A, Yang R, Rösl F, Mier W, Riemer AB. Therapeutic vaccination using minimal HPV16 epitopes in a novel MHC-humanized murine HPV tumor model. Oncoimmunology 2018; 8:e1524694. [PMID: 30546964 DOI: 10.1080/2162402x.2018.1524694] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/21/2018] [Accepted: 09/12/2018] [Indexed: 10/28/2022] Open
Abstract
Therapeutic vaccination as a treatment option for HPV-induced cancers is actively pursued because the two HPV proteins E6 and E7 represent ideal targets for immunotherapy, as they are non-self and expressed in all tumor stages. MHC-humanized mice are valuable tools for the study of therapeutic cancer vaccines - given the availability of a suitable tumor model. Here, we present for the first time an HPV16 tumor model suitable for fully MHC-humanized A2.DR1 mice, PAP-A2 cells, which in contrast to existing HPV16 tumor models allows the exclusive study of HLA-A2- and DR1-mediated immune responses, without any interfering murine MHC-presented epitopes. We used several HPV16 epitopes that were shown to be presented on human cervical cancer cells by mass spectrometry for therapeutic anti-tumor vaccination in the new tumor model. All epitopes were immunogenic when rendered amphiphilic by incorporation into a molecule containing stearic acids. Prophylactic and therapeutic vaccination experiments with the epitope E7/11-19 demonstrated that effective immune responses could be induced with these vaccination approaches in A2.DR1 mice. Interestingly, the combination of E7/11-19 with other immunogenic HPV16 E6/E7 epitopes caused a reduction of vaccine efficacy, although all tested combinations resulted in a survival benefit. In summary, we present the first HPV16 tumor model for exclusive studies of HLA-A2-mediated anti-HPV tumor immune responses and show anti-tumor efficacy of minimal epitope vaccines.
Collapse
Affiliation(s)
- Sebastian Kruse
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Marleen Büchler
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Philipp Uhl
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Max Sauter
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Scherer
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tammy C T Lan
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Samantha Zottnick
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alexandra Klevenz
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ruwen Yang
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Rösl
- Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Angelika B Riemer
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
11
|
Steinbach A, Winter J, Reuschenbach M, Blatnik R, Klevenz A, Bertrand M, Hoppe S, von Knebel Doeberitz M, Grabowska AK, Riemer AB. ERAP1 overexpression in HPV-induced malignancies: A possible novel immune evasion mechanism. Oncoimmunology 2017; 6:e1336594. [PMID: 28811980 DOI: 10.1080/2162402x.2017.1336594] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 01/17/2023] Open
Abstract
Immune evasion of tumors poses a major challenge for immunotherapy. For human papillomavirus (HPV)-induced malignancies, multiple immune evasion mechanisms have been described, including altered expression of antigen processing machinery (APM) components. These changes can directly influence epitope presentation and thus T-cell responses against tumor cells. To date, the APM had not been studied systematically in a large array of HPV+ tumor samples. Therefore in this study, systematic expression analysis of the APM was performed on the mRNA and protein level in a comprehensive collection of HPV16+ cell lines. Subsequently, HPV+ cervical tissue samples were examined by immunohistochemistry. ERAP1 (endoplasmic reticulum aminopeptidase 1) was the only APM component consistently altered - namely overexpressed - in HPV16+ tumor cell lines. ERAP1 was also found to be overexpressed in cervical intraepithelial neoplasia and cervical cancer samples; expression levels were increasing with disease stage. On the functional level, the influence of ERAP1 expression levels on HPV16 E7-derived epitope presentation was investigated by mass spectrometry and in cytotoxicity assays with HPV16-specific T-cell lines. ERAP1 overexpression did not cause a complete destruction of any of the HPV epitopes analyzed, however, an influence of ERAP1 overexpression on the presentation levels of certain HPV epitopes could be demonstrated by HPV16-specific CD8+ T-cells. These showed enhanced killing toward HPV16+ CaSki cells whose ERAP1 expression had been attenuated to normal levels. ERAP1 overexpression may thus represent a novel immune evasion mechanism in HPV-induced malignancies, in cases when presentation of clinically relevant epitopes is reduced by overactivity of this peptidase.
Collapse
Affiliation(s)
- Alina Steinbach
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Jan Winter
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miriam Reuschenbach
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Renata Blatnik
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Alexandra Klevenz
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miriam Bertrand
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie Hoppe
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | | | - Agnieszka K Grabowska
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angelika B Riemer
- Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
12
|
Dagvadorj N, Deuretzbacher A, Weisenberger D, Baumeister E, Trebing J, Lang I, Köchel C, Kapp M, Kapp K, Beilhack A, Hünig T, Einsele H, Wajant H, Grigoleit GU. Targeting of the WT1 91-138 fragment to human dendritic cells improves leukemia-specific T-cell responses providing an alternative approach to WT1-based vaccination. Cancer Immunol Immunother 2017; 66:319-332. [PMID: 27896368 PMCID: PMC11028450 DOI: 10.1007/s00262-016-1938-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 11/19/2016] [Indexed: 12/22/2022]
Abstract
Due to its immunogenicity and overexpression concomitant with leukemia progression, Wilms tumor protein 1 (WT1) is of particular interest for immunotherapy of AML relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT). So far, WT1-specific T-cell responses have mainly been induced by vaccination with peptides presented by certain HLA alleles. However, this approach is still not widely applicable in clinical practice due to common limitations of HLA restriction. Dendritic cell (DC) vaccines electroporated with mRNA encoding full-length protein have also been tested for generating WT1-derived peptides for presentation to T-cells. Alternatively, an efficient and broad WT1 peptide presentation could be elicited by triggering receptor-mediated protein endocytosis of DCs. Therefore, we developed antibody fusion proteins consisting of an antibody specific for the DEC205 endocytic receptor on human DCs and various fragments of WT1 as DC-targeting recombinant WT1 vaccines (anti-hDEC205-WT1). Of all anti-hDEC205-WT1 fusion proteins designed for overcoming insufficient expression, anti-hDEC205-WT110-35, anti-hDEC205-WT191-138, anti-hDEC205-WT1223-273, and anti-hDEC205-WT1324-371 were identified in good yields. The anti-hDEC205-WT191-138 was capable of directly inducing ex vivo T-cell responses by co-incubation of the fusion protein-loaded monocyte-derived mature DCs and autologous T-cells of either healthy or HSCT individuals. Furthermore, the DC-targeted WT191-138-induced specific T-cells showed a strong cytotoxic activity by lysing WT1-overexpressing THP-1 leukemia cells in vitro while sparing WT1-negative hematopoietic cells. In conclusion, our approach identifies four WT1 peptide-antibody fusion proteins with sufficient production and introduces an alternative vaccine that could be easily translated into clinical practice to improve WT1-directed antileukemia immune responses after allo-HSCT.
Collapse
Affiliation(s)
- Nergui Dagvadorj
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
- Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Anne Deuretzbacher
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Daniela Weisenberger
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Elke Baumeister
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Johannes Trebing
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Isabell Lang
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Carolin Köchel
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Markus Kapp
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Kerstin Kapp
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Andreas Beilhack
- Division of Experimental Stem Cell Transplantation, Interdisciplinary Center for Clinical Research, University of Würzburg, Würzburg, Germany
| | - Thomas Hünig
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Götz Ulrich Grigoleit
- Laboratory for Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany.
| |
Collapse
|
13
|
Nelde A, Walz JS, Kowalewski DJ, Schuster H, Wolz OO, Peper JK, Cardona Gloria Y, Langerak AW, Muggen AF, Claus R, Bonzheim I, Fend F, Salih HR, Kanz L, Rammensee HG, Stevanović S, Weber ANR. HLA class I-restricted MYD88 L265P-derived peptides as specific targets for lymphoma immunotherapy. Oncoimmunology 2016; 6:e1219825. [PMID: 28405493 PMCID: PMC5384368 DOI: 10.1080/2162402x.2016.1219825] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 12/28/2022] Open
Abstract
Genome sequencing has uncovered an array of recurring somatic mutations in different non-Hodgkin lymphoma (NHL) subtypes. If affecting protein-coding regions, such mutations may yield mutation-derived peptides that may be presented by HLA class I proteins and recognized by cytotoxic T cells. A recurring somatic and oncogenic driver mutation of the Toll-like receptor adaptor protein MYD88, Leu265Pro (L265P) was identified in up to 90% of different NHL subtype patients. We therefore screened the potential of MYD88L265P-derived peptides to elicit cytotoxic T cell responses as tumor-specific neoantigens. Based on in silico predictions, we identified potential MYD88L265P-containing HLA ligands for several HLA class I restrictions. A set of HLA class I MYD88L265P-derived ligands elicited specific cytotoxic T cell responses for HLA-B*07 and -B*15. These data highlight the potential of MYD88L265P mutation-specific peptide-based immunotherapy as a novel personalized treatment approach for patients with MYD88L265P+ NHLs that may complement pharmacological approaches targeting oncogenic MyD88 L265P signaling.
Collapse
Affiliation(s)
- Annika Nelde
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Juliane Sarah Walz
- Department of Hematology and Oncology, University Hospital Tübingen, Tübingen, Germany
| | | | - Heiko Schuster
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Olaf-Oliver Wolz
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Janet Kerstin Peper
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Yamel Cardona Gloria
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Anton W. Langerak
- Department of Immunology, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Alice F. Muggen
- Department of Immunology, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Rainer Claus
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, Germany
| | - Irina Bonzheim
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Falko Fend
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Helmut Rainer Salih
- Department of Hematology and Oncology, University Hospital Tübingen, Tübingen, Germany
- Clinical Cooperation Unit Translational Immunology, German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
| | - Lothar Kanz
- Department of Hematology and Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
| | - Alexander N. R. Weber
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
14
|
Jing L, Laing KJ, Dong L, Russell RM, Barlow RS, Haas JG, Ramchandani MS, Johnston C, Buus S, Redwood AJ, White KD, Mallal SA, Phillips EJ, Posavad CM, Wald A, Koelle DM. Extensive CD4 and CD8 T Cell Cross-Reactivity between Alphaherpesviruses. THE JOURNAL OF IMMUNOLOGY 2016; 196:2205-2218. [PMID: 26810224 DOI: 10.4049/jimmunol.1502366] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/15/2015] [Indexed: 12/18/2022]
Abstract
The Alphaherpesvirinae subfamily includes HSV types 1 and 2 and the sequence-divergent pathogen varicella zoster virus (VZV). T cells, controlled by TCR and HLA molecules that tolerate limited epitope amino acid variation, might cross-react between these microbes. We show that memory PBMC expansion with either HSV or VZV enriches for CD4 T cell lines that recognize the other agent at the whole-virus, protein, and peptide levels, consistent with bidirectional cross-reactivity. HSV-specific CD4 T cells recovered from HSV-seronegative persons can be explained, in part, by such VZV cross-reactivity. HSV-1-reactive CD8 T cells also cross-react with VZV-infected cells, full-length VZV proteins, and VZV peptides, as well as kill VZV-infected dermal fibroblasts. Mono- and cross-reactive CD8 T cells use distinct TCRB CDR3 sequences. Cross-reactivity to VZV is reconstituted by cloning and expressing TCRA/TCRB receptors from T cells that are initially isolated using HSV reagents. Overall, we define 13 novel CD4 and CD8 HSV-VZV cross-reactive epitopes and strongly imply additional cross-reactive peptide sets. Viral proteins can harbor both CD4 and CD8 HSV/VZV cross-reactive epitopes. Quantitative estimates of HSV/VZV cross-reactivity for both CD4 and CD8 T cells vary from 10 to 50%. Based on these findings, we hypothesize that host herpesvirus immune history may influence the pathogenesis and clinical outcome of subsequent infections or vaccinations for related pathogens and that cross-reactive epitopes and TCRs may be useful for multi-alphaherpesvirus vaccine design and adoptive cellular therapy.
Collapse
Affiliation(s)
- Lichen Jing
- Department of Medicine, University of Washington, Seattle, USA
| | - Kerry J Laing
- Department of Medicine, University of Washington, Seattle, USA
| | - Lichun Dong
- Department of Medicine, University of Washington, Seattle, USA
| | | | - Russell S Barlow
- Department of Global Health, University of Washington, Seattle, USA
| | - Juergen G Haas
- Max von Pettenkofer-Institute, Munich, Germany.,Division of Pathway Medicine, University of Edinburgh, United Kingdom
| | | | | | - Soren Buus
- Laboratory of Experimental Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Alec J Redwood
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia
| | - Katie D White
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
| | - Simon A Mallal
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
| | - Elizabeth J Phillips
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
| | - Christine M Posavad
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Department of Laboratory Medicine, University of Washington, Seattle, USA
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle, USA.,Department of Epidemiology, University of Washington, Seattle, USA.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Department of Laboratory Medicine, University of Washington, Seattle, USA
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, USA.,Department of Global Health, University of Washington, Seattle, USA.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Department of Laboratory Medicine, University of Washington, Seattle, USA.,Benaroya Research Institute, Seattle, USA
| |
Collapse
|
15
|
Caruso HG, Hurton LV, Najjar A, Rushworth D, Ang S, Olivares S, Mi T, Switzer K, Singh H, Huls H, Lee DA, Heimberger AB, Champlin RE, Cooper LJN. Tuning Sensitivity of CAR to EGFR Density Limits Recognition of Normal Tissue While Maintaining Potent Antitumor Activity. Cancer Res 2015; 75:3505-18. [PMID: 26330164 DOI: 10.1158/0008-5472.can-15-0139] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Many tumors overexpress tumor-associated antigens relative to normal tissue, such as EGFR. This limits targeting by human T cells modified to express chimeric antigen receptors (CAR) due to potential for deleterious recognition of normal cells. We sought to generate CAR(+) T cells capable of distinguishing malignant from normal cells based on the disparate density of EGFR expression by generating two CARs from monoclonal antibodies that differ in affinity. T cells with low-affinity nimotuzumab-CAR selectively targeted cells overexpressing EGFR, but exhibited diminished effector function as the density of EGFR decreased. In contrast, the activation of T cells bearing high-affinity cetuximab-CAR was not affected by the density of EGFR. In summary, we describe the generation of CARs able to tune T-cell activity to the level of EGFR expression in which a CAR with reduced affinity enabled T cells to distinguish malignant from nonmalignant cells.
Collapse
Affiliation(s)
- Hillary G Caruso
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Lenka V Hurton
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Amer Najjar
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Rushworth
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Sonny Ang
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simon Olivares
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tiejuan Mi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kirsten Switzer
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Harjeet Singh
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Helen Huls
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dean A Lee
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laurence J N Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas.
| |
Collapse
|
16
|
Antigenically Modified Human Pluripotent Stem Cells Generate Antigen-Presenting Dendritic Cells. Sci Rep 2015; 5:15262. [PMID: 26471005 PMCID: PMC4608011 DOI: 10.1038/srep15262] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/21/2015] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) provide a promising platform to produce dendritic cell (DC) vaccine. To streamline the production process, we investigated a unique antigen-loading strategy that suits this novel platform. Specifically, we stably modified hPSCs using tumour antigen genes in the form of a full-length tumour antigen gene or an artificial tumour antigen epitope-coding minigene. Such antigenically modified hPSCs were able to differentiate into tumour antigen-presenting DCs. Without conventional antigen-loading, DCs derived from the minigene-modified hPSCs were ready to prime a tumour antigen-specific T cell response and further expand these specific T cells in restimulation processes. These expanded tumour antigen-specific T cells were potent effectors with central memory or effector memory phenotype. Thus, we demonstrated that immunocompetent tumour antigen-loaded DCs can be directly generated from antigenically modified hPSCs. Using such strategy, we can completely eliminate the conventional antigen-loading step and significantly simplify the production of DC vaccine from hPSCs.
Collapse
|
17
|
Broadening the repertoire of melanoma-associated T-cell epitopes. Cancer Immunol Immunother 2015; 64:609-20. [PMID: 25854582 PMCID: PMC4412285 DOI: 10.1007/s00262-015-1664-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 02/02/2015] [Indexed: 12/13/2022]
Abstract
Immune therapy has provided a significant breakthrough in the treatment of metastatic melanoma. Despite the remarkable clinical efficacy and established involvement of effector CD8 T cells, the knowledge of the exact peptide-MHC complexes recognized by T cells on the tumor cell surface is limited. Many melanoma-associated T-cell epitopes have been described, but this knowledge remains largely restricted to HLA-A2, and we lack understanding of the T-cell recognition in the context of other HLA molecules. We selected six melanoma-associated antigens (MAGE-A3, NY-ESO-1, gp100, Mart1, tyrosinase and TRP-2) that are frequently recognized in patients with the aim of identifying novel T-cell epitopes restricted to HLA-A1, -A3, -A11 and -B7. Using in silico prediction and in vitro confirmation, we identified 127 MHC ligands and analyzed the T-cell responses against these ligands via the MHC multimer-based enrichment of peripheral blood from 39 melanoma patients and 10 healthy donors. To dissect the T-cell reactivity against this large peptide library, we used combinatorial-encoded MHC multimers and observed the T-cell responses against 17 different peptide-MHC complexes in the patient group and four in the healthy donor group. We confirmed the processing and presentation of HLA-A3-restricted T-cell epitopes from tyrosinase (TQYESGSMDK) and gp100 (LIYRRRLMK) and an HLA-A11-restricted T-cell epitope from gp100 (AVGATKVPR) via the cytolytic T-cell recognition of melanoma cell lines and/or K562 cells expressing the appropriate antigen and HLA molecule. We further found T-cell reactivity against two of the identified sequences among tumor-infiltrating lymphocytes from melanoma patients, suggesting a potential clinical relevance of these sequences.
Collapse
|
18
|
5-Azacytidine treatment sensitizes tumor cells to T-cell mediated cytotoxicity and modulates NK cells in patients with myeloid malignancies. Blood Cancer J 2014; 4:e197. [PMID: 24681961 PMCID: PMC3972700 DOI: 10.1038/bcj.2014.14] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/11/2014] [Indexed: 01/16/2023] Open
Abstract
Treatment with the demethylating agent 5-Azacytidine leads to prolonged survival for patients with myelodysplastic syndrome, and the demethylation induces upregulation of cancer-testis antigens. Cancer-testis antigens are well-known targets for immune recognition in cancer, and the immune system may have a role in this treatment regimen. We show here that 5-Azacytidine treatment leads to increased T-cell recognition of tumor cells. T-cell responses against a large panel of cancer-testis antigens were detected before treatment, and these responses were further induced upon initiation of treatment. These characteristics point to an ideal combination of 5-Azacytidine and immune therapy to preferentially boost T-cell responses against cancer-testis antigens. To initiate such combination therapy, essential knowledge is required about the general immune modulatory effect of 5-Azacytidine. We therefore examined potential treatment effects on both immune stimulatory (CD8 and CD4 T cells and Natural Killer (NK) cells) and immune inhibitory cell subsets (myeloid-derived suppressor cells and regulatory T cells). We observed a minor decrease and modulation of NK cells, but for all other populations no effects could be detected. Together, these data support a strategy for combining 5-Azacytidine treatment with immune therapy for potential clinical benefit.
Collapse
|
19
|
Lyngaa R, Pedersen NW, Schrama D, Thrue CA, Ibrani D, Met O, Thor Straten P, Nghiem P, Becker JC, Hadrup SR. T-cell responses to oncogenic merkel cell polyomavirus proteins distinguish patients with merkel cell carcinoma from healthy donors. Clin Cancer Res 2014; 20:1768-78. [PMID: 24526738 DOI: 10.1158/1078-0432.ccr-13-2697] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Merkel cell carcinoma (MCC) is a highly aggressive skin cancer with strong evidence of viral carcinogenesis. The association of MCC with the Merkel cell polyomavirus (MCPyV) may explain the explicit immunogenicity of MCC. Indeed, MCPyV-encoded proteins are likely targets for cytotoxic immune responses to MCC as they are both foreign to the host and necessary to maintain the oncogenic phenotype. However, to date only a single MCPyV-derived CD8 T-cell epitope has been described, thus impeding specific monitoring of T-cell responses to MCC. METHOD To overcome this limitation, we scanned the MCPyV oncoprotein large T and small T antigens and the virus capsid protein VP1 for potential T-cell epitopes, and tested for MHC class I affinity. We confirmed the relevance of these epitopes using a high-throughput platform for T-cell enrichment and combinatorial encoding of MHC class I multimers. RESULTS In peripheral blood from 38 patients with MCC and 30 healthy donors, we identified 53 MCPyV-directed CD8 T-cell responses against 35 different peptide sequences. Strikingly, T-cell responses against oncoproteins were exclusively present in patients with MCC, but not in healthy donors. We further demonstrate both the processing and presentation of the oncoprotein-derived epitopes, as well as the lytic activity of oncoprotein-specific T cells toward MHC-matched MCC cells. Demonstrating the presence of oncoprotein-specific T cells among tumor-infiltrating lymphocytes further substantiated the relevance of the identified epitopes. CONCLUSION These T-cell epitopes represent ideal targets for antigen-specific immune therapy of MCC, and enable tracking and characterization of MCPyV-specific immune responses.
Collapse
Affiliation(s)
- Rikke Lyngaa
- Authors' Affiliations: Center for Cancer Immune Therapy (CCIT), Department of Hematology, University Hospital Herlev, Herlev, Denmark; General Dermatology, Medical University of Graz, Graz, Austria; Departments of Medicine/Dermatology, Pathology, University of Washington; Fred Hutchinson Cancer Research Center, Seattle Cancer Center Care Alliance, Seattle, Washington; and Department of Oncology, University Hospital Herlev, Herlev, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Tong L, Schuhmacher C, Assenmacher M, Zänker K, Jähn P. Multiplex and functional detection of antigen-specific human T cells by ITRA--indirect T cell recognition assay. J Immunol Methods 2014; 404:13-23. [PMID: 24333463 DOI: 10.1016/j.jim.2013.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 08/14/2013] [Accepted: 11/19/2013] [Indexed: 12/24/2022]
Abstract
The identification and functional characterization of pathogen-specific T cells plays a critical role in immunological research and diagnostics. In addition to the present standard technologies such as intracellular cytokine staining (ICS), enzyme-linked immunospot (ELISPOT) and peptide-major-histocompatibility-complex (MHC) multimer staining, we aimed to develop a multiplex detection assay, which provides fast in vitro functional data for both human CD4 and CD8 T cells with different antigen specificities in one sample. In this study, we have exploited the expression of CD83 on B cells to develop the cell array-based indirect T cell recognition assay (ITRA). In detail, B cells are pulsed with different pathogen peptide pools and fluorescently barcoded. Thereafter the B cells are pooled and co-cultured with autologous T cells. Subsequently each B cell population is analyzed via flow cytometry for CD83 expression, which indicates antigen-specific interaction with CD4 T cells. Moreover, we revealed donor dependent variations of cytotoxic activity of pathogen-specific CD4 T cells and CD8 T cells, evidenced by specific lysis of peptide-pulsed B cells. Taken together, ITRA is a novel antigen presenting cell (APC) array based method to analyze the presence and function of various antigen-specific T cells in one sample. It has the potential to be used in the future for epitope/antigen screening in research and for analysis of anti-tumor, anti-pathogen or autoimmune T cell responses in patient samples.
Collapse
Affiliation(s)
- Lan Tong
- Department of Human Medicine, Institute of Immunology, University Witten/Herdecke, 58453 Witten, Germany
| | - Carolin Schuhmacher
- Department of Research and Development, Miltenyi Biotec GmbH, Bergisch Gladbach 51429, Germany
| | - Mario Assenmacher
- Department of Research and Development, Miltenyi Biotec GmbH, Bergisch Gladbach 51429, Germany
| | - Kurt Zänker
- Department of Human Medicine, Institute of Immunology, University Witten/Herdecke, 58453 Witten, Germany
| | - Peter Jähn
- Department of Research and Development, Miltenyi Biotec GmbH, Bergisch Gladbach 51429, Germany
| |
Collapse
|
21
|
Liao T, Kaufmann AM, Qian X, Sangvatanakul V, Chen C, Kube T, Zhang G, Albers AE. Susceptibility to cytotoxic T cell lysis of cancer stem cells derived from cervical and head and neck tumor cell lines. J Cancer Res Clin Oncol 2013; 139:159-70. [PMID: 23001491 DOI: 10.1007/s00432-012-1311-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 09/05/2012] [Indexed: 12/27/2022]
Abstract
PURPOSE To explore cancer stem cell susceptibility to a host's cytotoxic T lymphocyte (CTL)-mediated immune response. METHODS We compared the susceptibility of putative CSC generated from cancer cell lines to immunologic recognition and killing by alloantigen-specific CD8(+) CTL. CSC-enriched spheroid culture-derived cells (SDC) exhibited higher expression of ALDH, ICAM1 and of stem/progenitor cell markers on all 3 tumor cell lines investigated and lower MHC class I on the cervical cancer cell line as compared to their monolayer-derived cells (MDC). RESULTS The expression of ICAM1 and MHCI was upregulated by IFN-γ treatment. CSC populations were less sensitive to MHC class I-restricted alloantigen-specific CD8(+) CTL lysis as compared to matched MDC. IFN-γ pretreatment resulted in over-proportionally enhanced lysis of SDC. Finally, the subset of ALDH(high) expressing SDC presented more sensitivity toward CD8(+) CTL killing than the ALDH(low) SDC. CONCLUSIONS Tumor therapy resistance has been attributed to cancer stem cells (CSC). We show in vitro susceptibility of CSC to CTL-mediated lysis. Immunotherapy targeting of ALDH(+) CSC may therefore be a promising approach. Our results and method may be helpful for the development and optimization of adjuvants, as here exemplified for INF-γ, for CSC-targeted vaccines, independent of the availability of CSC-specific antigens.
Collapse
MESH Headings
- Aldehyde Dehydrogenase 1 Family
- B7-1 Antigen/analysis
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/therapy
- Cell Line, Tumor/immunology
- Cytotoxicity, Immunologic
- Female
- Flow Cytometry
- Gene Expression Regulation, Neoplastic
- Genes, MHC Class I
- Genes, MHC Class II
- Head and Neck Neoplasms/immunology
- Head and Neck Neoplasms/pathology
- Head and Neck Neoplasms/therapy
- Homeodomain Proteins/metabolism
- Humans
- Intercellular Adhesion Molecule-1/metabolism
- Interferon-gamma/pharmacology
- Isoenzymes/metabolism
- Male
- Nanog Homeobox Protein
- Neoplastic Stem Cells/immunology
- Octamer Transcription Factor-3/metabolism
- Retinal Dehydrogenase/metabolism
- SOXB1 Transcription Factors/metabolism
- Spheroids, Cellular
- T-Lymphocytes, Cytotoxic/immunology
- Uterine Cervical Neoplasms/immunology
- Uterine Cervical Neoplasms/pathology
- Uterine Cervical Neoplasms/therapy
Collapse
Affiliation(s)
- Tian Liao
- Clinic for Gynecology, Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|