51
|
Janelle V, Rulleau C, Del Testa S, Carli C, Delisle JS. T-Cell Immunotherapies Targeting Histocompatibility and Tumor Antigens in Hematological Malignancies. Front Immunol 2020; 11:276. [PMID: 32153583 PMCID: PMC7046834 DOI: 10.3389/fimmu.2020.00276] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Over the last decades, T-cell immunotherapy has revealed itself as a powerful, and often curative, strategy to treat blood cancers. In hematopoietic cell transplantation, most of the so-called graft-vs.-leukemia (GVL) effect hinges on the recognition of histocompatibility antigens that reflect immunologically relevant genetic variants between donors and recipients. Whether other variants acquired during the neoplastic transformation, or the aberrant expression of gene products can yield antigenic targets of similar relevance as the minor histocompatibility antigens is actively being pursued. Modern genomics and proteomics have enabled the high throughput identification of candidate antigens for immunotherapy in both autologous and allogeneic settings. As such, these major histocompatibility complex-associated tumor-specific (TSA) and tumor-associated antigens (TAA) can allow for the targeting of multiple blood neoplasms, which is a limitation for other immunotherapeutic approaches, such as chimeric antigen receptor (CAR)-modified T cells. We review the current strategies taken to translate these discoveries into T-cell therapies and propose how these could be introduced in clinical practice. Specifically, we discuss the criteria that are used to select the antigens with the greatest therapeutic value and we review the various T-cell manufacturing approaches in place to either expand antigen-specific T cells from the native repertoire or genetically engineer T cells with minor histocompatibility antigen or TSA/TAA-specific recombinant T-cell receptors. Finally, we elaborate on the current and future incorporation of these therapeutic T-cell products into the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Valérie Janelle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Caroline Rulleau
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Simon Del Testa
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Cédric Carli
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada.,Division Hématologie et Oncologie, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| |
Collapse
|
52
|
Kumbhari A, Kim PS, Lee PP. Optimisation of anti-cancer peptide vaccines to preferentially elicit high-avidity T cells. J Theor Biol 2020; 486:110067. [DOI: 10.1016/j.jtbi.2019.110067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/24/2019] [Accepted: 11/01/2019] [Indexed: 10/25/2022]
|
53
|
Vandamme C, Xicluna R, Hesnard L, Devaux M, Jaulin N, Guilbaud M, Le Duff J, Couzinié C, Moullier P, Saulquin X, Adjali O. Tetramer-Based Enrichment of Preexisting Anti-AAV8 CD8 + T Cells in Human Donors Allows the Detection of a T EMRA Subpopulation. Front Immunol 2020; 10:3110. [PMID: 32038634 PMCID: PMC6990124 DOI: 10.3389/fimmu.2019.03110] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/20/2019] [Indexed: 12/26/2022] Open
Abstract
Pre-existing immunity to AAV capsid may compromise the safety and efficiency of rAAV-mediated gene transfer in patients. Anti-capsid cytotoxic immune responses have proven to be a challenge to characterize because of the scarcity of circulating AAV-specific CD8+ T lymphocytes which can seldom be detected with conventional flow cytometry or ELISpot assays. Here, we used fluorescent MHC class I tetramers combined with magnetic enrichment to detect and phenotype AAV8-specific CD8+ T cells in human PBMCs without prior amplification. We showed that all healthy individuals tested carried a pool of AAV8-specific CD8+ T cells with a CD45RA+ CCR7- terminally-differentiated effector memory cell (TEMRA) fraction. Ex vivo frequencies of total AAV-specific CD8+ T cells were not predictive of IFNγ ELISpot responses but interestingly we evidenced a correlation between the proportion of TEMRA cells and IFNγ ELISpot positive responses. TEMRA cells may then play a role in recombinant AAV-mediated cytotoxicity in patients with preexisting immunity. Overall, our results encourage the development of new methods combining increased detection sensitivity of AAV-specific T cells and their poly-functional assessment to better characterize and monitor AAV capsid-specific cellular immune responses in the perspective of rAAV-mediated clinical trials.
Collapse
Affiliation(s)
- Céline Vandamme
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Rebecca Xicluna
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Leslie Hesnard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marie Devaux
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Nicolas Jaulin
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Mickaël Guilbaud
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Johanne Le Duff
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Célia Couzinié
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Philippe Moullier
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Xavier Saulquin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Oumeya Adjali
- INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| |
Collapse
|
54
|
Holmström MO, Cordua S, Skov V, Kjær L, Pallisgaard N, Ellervik C, Hasselbalch HC, Andersen MH. Evidence of immune elimination, immuno-editing and immune escape in patients with hematological cancer. Cancer Immunol Immunother 2020; 69:315-324. [PMID: 31915854 DOI: 10.1007/s00262-019-02473-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/31/2019] [Indexed: 10/25/2022]
Abstract
There is mounting evidence that the immune system can spontaneously clear malignant lesions before they manifest as overt cancer, albeit this activity has been difficult to demonstrate in humans. The calreticulin (CALR) exon 9 mutations are driver mutations in patients with chronic myeloproliferative neoplasms (MPN), which are chronic blood cancers. The CALR mutations generate a neo-antigen that is recognized by patient T cells, and T cells isolated from a patient with a CALR-mutation can recognize and kill autologous CALR-mutant cells. Surprisingly, healthy individuals display frequent and strong T cell responses to the CALR neo-antigens too. Furthermore, healthy individuals display immune responses to all parts of the mutant CALR epitope, and the CALR neo-epitope specific responses are memory T cell responses. These data suggest that although healthy individuals might acquire a CALR mutation, the mutant cells can be eliminated by the immune system. Additionally, a small fraction of healthy individuals harbor a CALR exon 9 mutation. Four healthy individuals carrying CALR mutations underwent a full medical examination including a bone marrow biopsy after a median follow up of 6.2 years. None of these patients displayed any signs of CALR-mutant MPN. Additionally, all healthy individuals displayed strong CALR neo-epitope specific T cell responses suggesting that these healthy individuals retained their CALR-mutant cells in the editing stage for several years. Thus, we suggest that CALR-mutant MPN could be a disease model of cancer immuno-editing, as we have demonstrated that CALR-mutant MPN displays all three stages described in the theory of cancer immuno-editing.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Herlev Hospital, Borgmester Ib Juuls Vej 25C, 5. Sal, 2730, Herlev, Denmark.
| | - Sabrina Cordua
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Niels Pallisgaard
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark.,Department of Laboratory Medicine, Harvard Medical School, Boston Children's Hospital, Boston, USA
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Herlev Hospital, Borgmester Ib Juuls Vej 25C, 5. Sal, 2730, Herlev, Denmark.,Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
55
|
Nagano S, Maeda T, Ichise H, Kashima S, Ohtaka M, Nakanishi M, Kitawaki T, Kadowaki N, Takaori-Kondo A, Masuda K, Kawamoto H. High Frequency Production of T Cell-Derived iPSC Clones Capable of Generating Potent Cytotoxic T Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 16:126-135. [PMID: 31970197 PMCID: PMC6965501 DOI: 10.1016/j.omtm.2019.12.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022]
Abstract
Current adoptive T cell therapies conducted in an autologous setting are costly, time-consuming, and depend on the quality of the patient’s T cells, and thus it would be highly beneficial to develop an allogeneic strategy. To this aim, we have developed a method by which cytotoxic T lymphocytes (CTLs) are regenerated from induced pluripotent stem cells that are originally derived from T cells (T-iPSCs). In order to assess the feasibility of this strategy, we investigated the frequency of usable T-iPSC clones in terms of their T cell-generating capability and T cell receptor (TCR) affinity. We first established eight clones of T-iPSCs bearing different MART-1-specific TCRs from a healthy volunteer. Whereas all clones were able to give rise to mature CTLs, cell yield varied greatly, and five clones were considered to be usable. TCR affinity in the regenerated CTLs showed a large variance among the eight clones, but functional avidities measured by cytotoxic activity were almost equivalent among three selected clones representing high, medium, and low TCR affinity. In a total of 50 alloreactivity tests using five CTL clones versus ten target cells, alloreactivity was seen in only three cases. These findings collectively support the feasibility of this T-iPSC strategy.
Collapse
Affiliation(s)
- Seiji Nagano
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takuya Maeda
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Ichise
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Soki Kashima
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Department of Urology, Akita University Graduate School of Medicine, Akita City, Japan
| | - Manami Ohtaka
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Toshio Kitawaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norimitsu Kadowaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kyoko Masuda
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Kawamoto
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Corresponding author: Hiroshi Kawamoto, Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
56
|
De Simone G, Mazza EMC, Cassotta A, Davydov AN, Kuka M, Zanon V, De Paoli F, Scamardella E, Metsger M, Roberto A, Pilipow K, Colombo FS, Tenedini E, Tagliafico E, Gattinoni L, Mavilio D, Peano C, Price DA, Singh SP, Farber JM, Serra V, Cucca F, Ferrari F, Orrù V, Fiorillo E, Iannacone M, Chudakov DM, Sallusto F, Lugli E. CXCR3 Identifies Human Naive CD8 + T Cells with Enhanced Effector Differentiation Potential. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:3179-3189. [PMID: 31740485 PMCID: PMC6900484 DOI: 10.4049/jimmunol.1901072] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/16/2019] [Indexed: 01/19/2023]
Abstract
In mice, the ability of naive T (TN) cells to mount an effector response correlates with TCR sensitivity for self-derived Ags, which can be quantified indirectly by measuring surface expression levels of CD5. Equivalent findings have not been reported previously in humans. We identified two discrete subsets of human CD8+ TN cells, defined by the absence or presence of the chemokine receptor CXCR3. The more abundant CXCR3+ TN cell subset displayed an effector-like transcriptional profile and expressed TCRs with physicochemical characteristics indicative of enhanced interactions with peptide-HLA class I Ags. Moreover, CXCR3+ TN cells frequently produced IL-2 and TNF in response to nonspecific activation directly ex vivo and differentiated readily into Ag-specific effector cells in vitro. Comparative analyses further revealed that human CXCR3+ TN cells were transcriptionally equivalent to murine CXCR3+ TN cells, which expressed high levels of CD5. These findings provide support for the notion that effector differentiation is shaped by heterogeneity in the preimmune repertoire of human CD8+ T cells.
Collapse
Affiliation(s)
- Gabriele De Simone
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Emilia M C Mazza
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, 8093 Zurich, Switzerland
| | - Alexey N Davydov
- Central European Institute of Technology, 621 00 Brno, Czech Republic
| | - Mirela Kuka
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS, San Raffaele Scientific Institute and Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Veronica Zanon
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Federica De Paoli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Eloise Scamardella
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Maria Metsger
- Central European Institute of Technology, 621 00 Brno, Czech Republic
| | - Alessandra Roberto
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Karolina Pilipow
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Federico S Colombo
- Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Elena Tenedini
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Enrico Tagliafico
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Luca Gattinoni
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
- Regensburg Center for Interventional Immunology, University Regensburg and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20122 Milan, Italy
| | - Clelia Peano
- Division of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy
- Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Satya P Singh
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Joshua M Farber
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | - Valeria Orrù
- IRGB, National Research Council, 09042 Monserrato, Italy
| | | | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS, San Raffaele Scientific Institute and Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Dmitriy M Chudakov
- Central European Institute of Technology, 621 00 Brno, Czech Republic
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; and
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Federica Sallusto
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, 8093 Zurich, Switzerland
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy;
- Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| |
Collapse
|
57
|
Richmond JM, Strassner JP, Zapata L, Garg M, Riding RL, Refat MA, Fan X, Azzolino V, Tovar-Garza A, Tsurushita N, Pandya AG, Tso JY, Harris JE. Antibody blockade of IL-15 signaling has the potential to durably reverse vitiligo. Sci Transl Med 2019; 10:10/450/eaam7710. [PMID: 30021889 DOI: 10.1126/scitranslmed.aam7710] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/26/2018] [Accepted: 06/14/2018] [Indexed: 01/18/2023]
Abstract
Vitiligo is an autoimmune disease of the skin mediated by CD8+ T cells that kill melanocytes and create white spots. Skin lesions in vitiligo frequently return after discontinuing conventional treatments, supporting the hypothesis that autoimmune memory is formed at these locations. We found that lesional T cells in mice and humans with vitiligo display a resident memory (TRM) phenotype, similar to those that provide rapid, localized protection against reinfection from skin and mucosal-tropic viruses. Interleukin-15 (IL-15)-deficient mice reportedly have impaired TRM formation, and IL-15 promotes TRM function ex vivo. We found that both human and mouse TRM express the CD122 subunit of the IL-15 receptor and that keratinocytes up-regulate CD215, the subunit required to display the cytokine on their surface to promote activation of T cells. Targeting IL-15 signaling with an anti-CD122 antibody reverses disease in mice with established vitiligo. Short-term treatment with anti-CD122 inhibits TRM production of interferon-γ (IFNγ), and long-term treatment depletes TRM from skin lesions. Short-term treatment with anti-CD122 can provide durable repigmentation when administered either systemically or locally in the skin. On the basis of these data, we propose that targeting CD122 may be a highly effective and even durable treatment strategy for vitiligo and other tissue-specific autoimmune diseases involving TRM.
Collapse
Affiliation(s)
- Jillian M Richmond
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - James P Strassner
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lucio Zapata
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Madhuri Garg
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Rebecca L Riding
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Maggi A Refat
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Xueli Fan
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Vincent Azzolino
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | - Amit G Pandya
- University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - J Yun Tso
- JN Biosciences LLC, Mountain View, CA 94043, USA
| | - John E Harris
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
58
|
Gerber HP, Sibener LV, Lee LJ, Gee M. Intracellular targets as source for cleaner targets for the treatment of solid tumors. Biochem Pharmacol 2019; 168:275-284. [DOI: 10.1016/j.bcp.2019.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/12/2019] [Indexed: 01/02/2023]
|
59
|
Burrack AL, Spartz EJ, Raynor JF, Wang I, Olson M, Stromnes IM. Combination PD-1 and PD-L1 Blockade Promotes Durable Neoantigen-Specific T Cell-Mediated Immunity in Pancreatic Ductal Adenocarcinoma. Cell Rep 2019; 28:2140-2155.e6. [PMID: 31433988 PMCID: PMC7975822 DOI: 10.1016/j.celrep.2019.07.059] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/17/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal cancer resistant to immunotherapy. We create a PDA mouse model and show that neoantigen expression is required for intratumoral T cell accumulation and response to immune checkpoint blockade. By generating a peptide:MHC tetramer, we identify that PDA induces rapid intratumoral, and progressive systemic, tumor-specific T cell exhaustion. Monotherapy PD-1 or PD-L1 blockade enhances systemic T cell expansion and induces objective responses that require systemic T cells. However, tumor escape variants defective in IFNγ-inducible Tap1 and MHC class I cell surface expression ultimately emerge. Combination PD-1 + PD-L1 blockade synergizes therapeutically by increasing intratumoral KLRG1+Lag3-TNFα+ tumor-specific T cells and generating memory T cells capable of expanding to spontaneous tumor recurrence, thereby prolonging animal survival. Our studies support that PD-1 and PD-L1 are relevant immune checkpoints in PDA and identify a combination for clinical testing in those patients with neoantigen-specific T cells.
Collapse
Affiliation(s)
- Adam L Burrack
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Ellen J Spartz
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jackson F Raynor
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Iris Wang
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Margaret Olson
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Ingunn M Stromnes
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Masonic Cancer Center of the University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
60
|
Pinho MP, Patente TA, Flatow EA, Sallusto F, Barbuto JAM. Frequency determination of breast tumor-reactive CD4 and CD8 T cells in humans: unveiling the antitumor immune response. Oncoimmunology 2019; 8:1607674. [PMID: 31413917 DOI: 10.1080/2162402x.2019.1607674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/21/2019] [Accepted: 04/07/2019] [Indexed: 01/11/2023] Open
Abstract
As cancer immunotherapy gains importance, the determination of a patient's ability to react to his/her tumor is unquestionably relevant. Though the presence of T cells that recognize specific tumor antigens is well established, the total frequency of tumor-reactive T cells in humans is difficult to assess, especially due to the lack of broad analysis techniques. Here, we describe a strategy that allows this determination, in both CD4 and CD8 compartments, using T cell proliferation induced by tumor cell-lysate pulsed dendritic cells as the readout. All 12 healthy donor tested had circulating CD4 and CD8 tumor cell-reactive T cells. The detection of these T cells, not only in the naïve but also in the memory compartment, can be seen as an evidence of tumor immunosurveillance in humans. As expected, breast cancer patients had higher frequencies of blood tumor-reactive T cells, but with differences among breast cancer subtypes. Interestingly, the frequency of blood tumor-reactive T cells in patients did not correlate to the frequency of infiltrating tumor-reactive T cells, highlighting the danger of implying a local tumor response from blood obtained data. In conclusion, these data add T cell evidence to immunosurveillance in humans, confirm that immune parameters in blood may be misleading and describe a tool to follow the tumor-specific immune response in patients and, thus, to design better immunotherapeutic approaches.
Collapse
Affiliation(s)
- Mariana Pereira Pinho
- Department of Immunology, Institute of Biomedical Sciences of the University of Sao Paulo, Sao Paulo, Brazil
| | - Thiago Andrade Patente
- Department of Immunology, Institute of Biomedical Sciences of the University of Sao Paulo, Sao Paulo, Brazil
| | - Elizabeth Alexandra Flatow
- Department of Immunology, Institute of Biomedical Sciences of the University of Sao Paulo, Sao Paulo, Brazil
| | - Federica Sallusto
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera italiana, Bellinzona, Switzerland.,Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
61
|
Induction of neoantigen-reactive T cells from healthy donors. Nat Protoc 2019; 14:1926-1943. [DOI: 10.1038/s41596-019-0170-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 03/21/2019] [Indexed: 12/21/2022]
|
62
|
Martinez-Usatorre A, Sempere LF, Carmona SJ, Carretero-Iglesia L, Monnot G, Speiser DE, Rufer N, Donda A, Zehn D, Jandus C, Romero P. MicroRNA-155 Expression Is Enhanced by T-cell Receptor Stimulation Strength and Correlates with Improved Tumor Control in Melanoma. Cancer Immunol Res 2019; 7:1013-1024. [PMID: 31043416 DOI: 10.1158/2326-6066.cir-18-0504] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/24/2018] [Accepted: 04/26/2019] [Indexed: 11/16/2022]
Abstract
microRNAs are short noncoding RNAs that regulate protein expression posttranscriptionally. We previously showed that miR-155 promotes effector CD8+ T-cell responses. However, little is known about the regulation of miR-155 expression. Here, we report that antigen affinity and dose determine miR-155 expression in CD8+ T cells. In B16 tumors expressing a low-affinity antigen ligand, tumor-specific infiltrating CD8+ T cells showed variable miR-155 expression, whereby high miR-155 expression was associated with more cytokine-producing cells and tumor control. Moreover, anti-PD-1 treatment led to both increased miR-155 expression and tumor control by specific CD8+ T cells. In addition, miR-155 overexpression enhanced exhausted CD8+ T-cell persistence in the LCMV cl13 chronic viral infection model. In agreement with these observations in mouse models, miR-155 expression in human effector memory CD8+ T cells positively correlated with their frequencies in tumor-infiltrated lymph nodes of melanoma patients. Low miR-155 target gene signature in tumors was associated with prolonged overall survival in melanoma patients. Altogether, these results raise the possibility that high miR-155 expression in CD8+ tumor-infiltrating T cells may be a surrogate marker of the relative potency of in situ antigen-specific CD8+ T-cell responses.
Collapse
Affiliation(s)
| | - Lorenzo F Sempere
- Department of Radiology, Precision Health Program, Michigan State University, East Lansing, Michigan
| | - Santiago J Carmona
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Laura Carretero-Iglesia
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Gwennaëlle Monnot
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Daniel E Speiser
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Nathalie Rufer
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Alena Donda
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Dietmar Zehn
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Camilla Jandus
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Pedro Romero
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
63
|
Przybyla A, Zhang T, Li R, Roen DR, Mackiewicz A, Lehmann PV. Natural T cell autoreactivity to melanoma antigens: clonally expanded melanoma-antigen specific CD8 + memory T cells can be detected in healthy humans. Cancer Immunol Immunother 2019; 68:709-720. [PMID: 30783693 PMCID: PMC11028361 DOI: 10.1007/s00262-018-02292-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 12/24/2018] [Indexed: 12/30/2022]
Abstract
We used four-color ImmunoSpot® assays, in conjunction with peptide pools that cover the sequence of tyrosinase (Tyr), melanoma-associated antigen A3 (MAGE-A3), melanocyte antigen/melanoma antigen recognized by T cells 1 (Melan-A/MART-1), glycoprotein 100 (gp100), and New York esophageal squamous cell carcinoma-1 (NY-ESO-1) to characterize the melanoma antigen (MA)-specific CD8 + cell repertoire in PBMC of 40 healthy human donors (HD). Tyr triggered interferon gamma (IFN-γ)-secreting CD8 + T cells in 25% of HD within 24 h of antigen stimulation ex vivo. MAGE-A3, Melan-A/MART-1, and gp100 also induced recall responses in 10%, 7.5%, and 2.5% of HD, respectively. At this time point, these CD8 + T cells did not yet produce GzB (granzyme B). However, they engaged in GzB production after 72 h of antigen stimulation. By this 72-h time point, 57.5% of the HD responded to at least one, and typically several, of the MA. A closer characterization of the Tyr-specific CD8 + T cell repertoire indicated that it was low-affinity, and to primarily entail a stem cell-like subpopulation. Collectively, our data reveal pre-existing endogenous T cell immunity against melanoma antigens in healthy donors, and analogous to natural autoantibodies, we have termed this "natural T cell autoreactivity".
Collapse
Affiliation(s)
- Anna Przybyla
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Ting Zhang
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
| | - Ruliang Li
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
| | - Diana R Roen
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Paul V Lehmann
- Research and Development Department, Cellular Technology Limited (CTL), 20521 Chagrin Boulevard, Shaker Heights, Cleveland, OH, 44122-5350, USA.
| |
Collapse
|
64
|
Holmström MO, Hasselbalch HC, Andersen MH. Neo-antigen specific memory T-cell responses in healthy individuals. Oncoimmunology 2019; 8:1599640. [PMID: 31143522 DOI: 10.1080/2162402x.2019.1599640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
The driver mutations in exon 9 of the calreticulin protein have only been identified in patients with myeloid cancers. We recently demonstrated that healthy individuals display strong and frequent T-cell responses towards this mutation. This memory T-cell response is likely evidence of the elimination of mutated cells in healthy individuals.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark.,National Center for Cancer Immune Therapy, Department of Hematology, Herlev University Hospital, Herlev, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Hematology, Herlev University Hospital, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
65
|
Gutjahr A, Papagno L, Nicoli F, Kanuma T, Kuse N, Cabral-Piccin MP, Rochereau N, Gostick E, Lioux T, Perouzel E, Price DA, Takiguchi M, Verrier B, Yamamoto T, Paul S, Appay V. The STING ligand cGAMP potentiates the efficacy of vaccine-induced CD8+ T cells. JCI Insight 2019; 4:125107. [PMID: 30944257 DOI: 10.1172/jci.insight.125107] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/14/2019] [Indexed: 11/17/2022] Open
Abstract
Pathogen recognition receptor (PRR) agonists are currently being developed and tested as adjuvants in various formulations to optimize the immunogenicity and efficacy of vaccines. Using an original in vitro approach to prime naive precursors from unfractionated human peripheral blood mononuclear cells, we assessed the influence of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), a ligand for the stimulator of interferon genes (STING), on the induction of antigen-specific CD8+ T cells. We found that 2'3'-cGAMP and 3'3'-cGAMP were especially potent adjuvants in this system, driving the expansion and maturation of functionally replete antigen-specific CD8+ T cells via the induction of type I IFNs. The biological relevance of these findings was confirmed in vivo using two mouse models, in which 2'3'-cGAMP-adjuvanted vaccination elicited protective antitumor or antiviral CD8+ T cell responses. These results identify particular isoforms of cGAMP as effective adjuvants that may find utility in the development of novel immunotherapies and vaccines.
Collapse
Affiliation(s)
- Alice Gutjahr
- Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM, Centre d'Investigation Clinique en Vaccinologie 1408, Faculté de Médecine de Saint-Etienne, Saint-Etienne, France.,Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Unité Mixte de Recherche 5305, Université Lyon 1, CNRS, Lyon, France.,InvivoGen, Toulouse, France
| | - Laura Papagno
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Francesco Nicoli
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Tomohiro Kanuma
- Laboratory of Immunosenescence, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Nozomi Kuse
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | | | | | - Emma Gostick
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | | | | | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | | | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Unité Mixte de Recherche 5305, Université Lyon 1, CNRS, Lyon, France
| | - Takuya Yamamoto
- Laboratory of Immunosenescence, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Stéphane Paul
- Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM, Centre d'Investigation Clinique en Vaccinologie 1408, Faculté de Médecine de Saint-Etienne, Saint-Etienne, France
| | - Victor Appay
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| |
Collapse
|
66
|
Richmond JM, Strassner JP, Rashighi M, Agarwal P, Garg M, Essien KI, Pell LS, Harris JE. Resident Memory and Recirculating Memory T Cells Cooperate to Maintain Disease in a Mouse Model of Vitiligo. J Invest Dermatol 2019; 139:769-778. [PMID: 30423329 PMCID: PMC6431571 DOI: 10.1016/j.jid.2018.10.032] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/24/2018] [Accepted: 10/28/2018] [Indexed: 10/27/2022]
Abstract
Tissue resident memory T cells (Trm) form in the skin in vitiligo and persist to maintain disease, as white spots often recur rapidly after discontinuing therapy. We and others have recently described melanocyte-specific autoreactive Trm in vitiligo lesions. Here, we characterize the functional relationship between Trm and recirculating memory T cells (Tcm) in our vitiligo mouse model. We found that both Trm and Tcm sensed autoantigen in the skin long after stabilization of disease, producing IFN-γ, CXCL9, and CXCL10. Blockade of Tcm recruitment to the skin with FTY720 or depletion of Tcm with low-dose Thy1.1 antibody reversed disease, indicating that Trm cooperate with Tcm to maintain disease. Taken together, our data provide characterization of skin memory T cells in vitiligo, demonstrate that Trm and Tcm work together during disease, and indicate that targeting their survival or function may provide novel, durable treatment options for patients.
Collapse
Affiliation(s)
- Jillian M Richmond
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - James P Strassner
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Mehdi Rashighi
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Priti Agarwal
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Madhuri Garg
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kingsley I Essien
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Lila S Pell
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - John E Harris
- Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| |
Collapse
|
67
|
Inhibition of Heme Oxygenase-1 Activity Enhances Wilms Tumor-1-Specific T-Cell Responses in Cancer Immunotherapy. Int J Mol Sci 2019; 20:ijms20030482. [PMID: 30678050 PMCID: PMC6387130 DOI: 10.3390/ijms20030482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023] Open
Abstract
Wilms tumor protein-1 (WT1) is an attractive target for adoptive T-cell therapy due to its expression in solid tumors and hematologic malignancies. However, T cells recognizing WT1 occur in low frequencies in the peripheral blood of healthy donors, limiting potential therapeutic possibilities. Tin mesoporphyrin (SnMP) is known to inhibit heme oxygenase-1 (HO-1), which has been shown to boost the activation and proliferation of human virus-specific T cells. We analyzed the influence of this effect on the generation of WT1-specific T cells and developed strategies for generating quantities of these cells from healthy donors, sufficient for adoptive T-cell therapies. HO-1 inhibition with SnMP increased WT1-specific T-cell frequencies in 13 (26%) of 50 healthy donors. To assess clinical applicability, we measured the enrichment efficiency of SnMP-treated WT1-specific T cells in response to a WT1-specific peptide pool and a HLA-A*02:01-restricted WT1 peptide by cytokine secretion assay. SnMP treatment resulted in a 28-fold higher enrichment efficacy with equal functionality. In conclusion, pharmacological inhibition of HO-1 activity with SnMP results in more efficient generation of functionally active WT1-specific T cells. This study demonstrates the therapeutic potentials of inhibiting HO-1 with SnMP to enhance antigen-specific T-cell responses in the treatment of cancer patients with WT1-positive disease.
Collapse
|
68
|
Holmström MO, Ahmad SM, Klausen U, Bendtsen SK, Martinenaite E, Riley CH, Svane IM, Kjær L, Skov V, Ellervik C, Pallisgaard N, Hasselbalch HC, Andersen MH. High frequencies of circulating memory T cells specific for calreticulin exon 9 mutations in healthy individuals. Blood Cancer J 2019; 9:8. [PMID: 30655510 PMCID: PMC6336769 DOI: 10.1038/s41408-018-0166-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
Mutations in exon 9 of the calreticulin gene (CALR) frequently occur in patients with chronic myeloproliferative neoplasms (MPN). Patients exhibit spontaneous cellular immune responses to epitopes derived from the mutant CALR C-terminus, and CALR-mutant-specific T cells recognize autologous CALR-mutant malignant cells. This study investigated whether CALR-mutant-specific T cells occur naturally in CALRwt MPN-patients and in healthy individuals. Specific immune responses against epitopes in the mutant CALR peptide sequence were detected in both CALRwt MPN-patients and in healthy individuals. Healthy donors displayed more frequent and stronger CALR-mutant specific T-cell responses compared to the responses identified in CALR-mutant MPN-patients. Several T-cell responses were identified in healthy donors directly ex vivo. Importantly, by running functional analyses on live-sorted immune cells from healthy donors, we showed that circulating CALR-mutant-specific immune cells are T-memory cells. These findings suggest, that healthy individuals acquire a CALR exon 9 mutation, but the immune system reacts and clears the mutant cells, and during this reaction generates CALR-mutant specific T-memory cells. We believe that these findings provide the evidence for tumor immune surveillance in MPN.
Collapse
Affiliation(s)
- Morten O Holmström
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark. .,Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Herlev, Denmark.
| | - Shamaila M Ahmad
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Uffe Klausen
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Simone K Bendtsen
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Evelina Martinenaite
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | | | - Inge M Svane
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Herlev, Denmark.,Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark.,Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Niels Pallisgaard
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Hans C Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Mads H Andersen
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
69
|
Brunner T. Ecto-calreticulin is essential for an efficient immunogenic cell death stimulation in mouse melanoma. Genes Immun 2019; 20:527-528. [DOI: 10.1038/s41435-018-0054-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
70
|
Löffler MW, Kowalewski DJ, Backert L, Bernhardt J, Adam P, Schuster H, Dengler F, Backes D, Kopp HG, Beckert S, Wagner S, Königsrainer I, Kohlbacher O, Kanz L, Königsrainer A, Rammensee HG, Stevanović S, Haen SP. Mapping the HLA Ligandome of Colorectal Cancer Reveals an Imprint of Malignant Cell Transformation. Cancer Res 2018; 78:4627-4641. [PMID: 29789417 DOI: 10.1158/0008-5472.can-17-1745] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/15/2017] [Accepted: 05/16/2018] [Indexed: 11/16/2022]
Abstract
Immune cell infiltrates have proven highly relevant for colorectal carcinoma prognosis, making colorectal cancer a promising candidate for immunotherapy. Because tumors interact with the immune system via HLA-presented peptide ligands, exact knowledge of the peptidome constitution is fundamental for understanding this relationship. Here, we comprehensively describe the naturally presented HLA ligandome of colorectal carcinoma and corresponding nonmalignant colon (NMC) tissue. Mass spectrometry identified 35,367 and 28,132 HLA class I ligands on colorectal carcinoma and NMC, attributable to 7,684 and 6,312 distinct source proteins, respectively. Cancer-exclusive peptides were assessed on source protein level using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein analysis through evolutionary relationships (PANTHER), revealing pathognomonic colorectal carcinoma-associated pathways, including Wnt, TGFβ, PI3K, p53, and RTK-RAS. Relative quantitation of peptide presentation on paired colorectal carcinoma and NMC tissue further identified source proteins from cancer- and infection-associated pathways to be overrepresented merely within the colorectal carcinoma ligandome. From the pool of tumor-exclusive peptides, a selected HLA-ligand subset was assessed for immunogenicity, with the majority exhibiting an existing T-cell repertoire. Overall, these data show that the HLA ligandome reflects cancer-associated pathways implicated in colorectal carcinoma oncogenesis, suggesting that alterations in tumor cell metabolism could result in cancer-specific, albeit not mutation-derived, tumor antigens. Hence, a defined pool of unique tumor peptides, attributable to complex cellular alterations that are exclusive to malignant cells, might comprise promising candidates for immunotherapeutic applications.Significance: Cancer-associated pathways are reflected in the antigenic landscape of colorectal cancer, suggesting that tumor-specific antigens do not necessarily have to be mutation-derived but may also originate from other alterations in cancer cells. Cancer Res; 78(16); 4627-41. ©2018 AACR.
Collapse
Affiliation(s)
- Markus W Löffler
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany.,University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Tübingen, Germany
| | - Daniel J Kowalewski
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Linus Backert
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany.,University of Tübingen, Center for Bioinformatics, Tübingen, Germany
| | - Jörg Bernhardt
- University of Greifswald, Institute of Microbiology, Greifswald, Germany
| | | | - Heiko Schuster
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Florian Dengler
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany.,University Hospital Tübingen, Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, Tübingen, Germany
| | - Daniel Backes
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany.,University Hospital Tübingen, Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, Tübingen, Germany
| | - Hans-Georg Kopp
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany.,University Hospital Tübingen, Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, Tübingen, Germany
| | - Stefan Beckert
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Tübingen, Germany
| | - Silvia Wagner
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Tübingen, Germany
| | - Ingmar Königsrainer
- University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Tübingen, Germany
| | - Oliver Kohlbacher
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany.,University of Tübingen, Center for Bioinformatics, Tübingen, Germany.,University of Tübingen, Quantitative Biology Center (QBiC), Tübingen, Germany.,Max Planck Institute for Developmental Biology, Biomolecular Interactions, Tübingen, Germany
| | - Lothar Kanz
- University Hospital Tübingen, Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, Tübingen, Germany
| | - Alfred Königsrainer
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany.,University Hospital Tübingen, Department of General, Visceral and Transplant Surgery, Tübingen, Germany
| | - Hans-Georg Rammensee
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany
| | - Sebastian P Haen
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany.,University Hospital Tübingen, Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, Tübingen, Germany
| |
Collapse
|
71
|
Mocellin S, Panelli M, Wang E, Rossi CR, Marincola FM. Tumor Microenvironment: What have we Learned Studying the Immune Response in this Puzzling Battlefield? TUMORI JOURNAL 2018; 88:437-44. [PMID: 12597134 DOI: 10.1177/030089160208800601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent developments hallmark the progress in the understanding of tumor immunology and related therapeutic strategies. The administration of interleukin-2 (IL-2) to patients with cancer has shown that immune manipulation can mediate the regression of established cancers. The identification of the genes encoding cancer antigens and the development of means for effectively immunizing against these antigens has opened new avenues for the development of active immunization of patients with cancer. However, an efficient immune response against tumor comprises an intricate molecular network still poorly understood. Only when the code governing immune responsiveness of cancer will be deciphered, new therapeutic strategies could be designed to fit biologically defined mechanisms of immune rejection of cancer. In this review, we propose that the mechanisms regulating tumor rejection in response to vaccination will be more efficiently identified by following the evolution of treatment induced events within the tumor microenvironment taking advantage of recently developed technological tools. As a model, we will discuss the observed immune response to tumor antigen -specific immunization and its relationship with the systemic administration of IL-2.
Collapse
Affiliation(s)
- Simone Mocellin
- Immunnogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
72
|
Crittenden MR, Baird J, Friedman D, Savage T, Uhde L, Alice A, Cottam B, Young K, Newell P, Nguyen C, Bambina S, Kramer G, Akporiaye E, Malecka A, Jackson A, Gough MJ. Mertk on tumor macrophages is a therapeutic target to prevent tumor recurrence following radiation therapy. Oncotarget 2018; 7:78653-78666. [PMID: 27602953 PMCID: PMC5346667 DOI: 10.18632/oncotarget.11823] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 08/25/2016] [Indexed: 12/19/2022] Open
Abstract
Radiation therapy provides a means to kill large numbers of cancer cells in a controlled location resulting in the release of tumor-specific antigens and endogenous adjuvants. However, by activating pathways involved in apoptotic cell recognition and phagocytosis, irradiated cancer cells engender suppressive phenotypes in macrophages. We demonstrate that the macrophage-specific phagocytic receptor, Mertk is upregulated in macrophages in the tumor following radiation therapy. Ligation of Mertk on macrophages results in anti-inflammatory cytokine responses via NF-kB p50 upregulation, which in turn limits tumor control following radiation therapy. We demonstrate that in immunogenic tumors, loss of Mertk is sufficient to permit tumor cure following radiation therapy. However, in poorly immunogenic tumors, TGFβ inhibition is also required to result in tumor cure following radiation therapy. These data demonstrate that Mertk is a highly specific target whose absence permits tumor control in combination with radiation therapy.
Collapse
Affiliation(s)
- Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA.,The Oregon Clinic, Portland OR, USA
| | - Jason Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - David Friedman
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Talicia Savage
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Lauren Uhde
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Alejandro Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Benjamin Cottam
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Kristina Young
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA.,The Oregon Clinic, Portland OR, USA
| | - Pippa Newell
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA.,Providence Hepatobiliary and Pancreatic Cancer Program, Providence Portland Medical Center, Portland OR, USA
| | - Cynthia Nguyen
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Gwen Kramer
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Emmanuel Akporiaye
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Anna Malecka
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, University of Nottingham, UK
| | - Andrew Jackson
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, University of Nottingham, UK
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| |
Collapse
|
73
|
Bovay A, Zoete V, Dolton G, Bulek AM, Cole DK, Rizkallah PJ, Fuller A, Beck K, Michielin O, Speiser DE, Sewell AK, Fuertes Marraco SA. T cell receptor alpha variable 12-2 bias in the immunodominant response to Yellow fever virus. Eur J Immunol 2018; 48:258-272. [PMID: 28975614 PMCID: PMC5887915 DOI: 10.1002/eji.201747082] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/15/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022]
Abstract
The repertoire of human αβ T-cell receptors (TCRs) is generated via somatic recombination of germline gene segments. Despite this enormous variation, certain epitopes can be immunodominant, associated with high frequencies of antigen-specific T cells and/or exhibit bias toward a TCR gene segment. Here, we studied the TCR repertoire of the HLA-A*0201-restricted epitope LLWNGPMAV (hereafter, A2/LLW) from Yellow Fever virus, which generates an immunodominant CD8+ T cell response to the highly effective YF-17D vaccine. We discover that these A2/LLW-specific CD8+ T cells are highly biased for the TCR α chain TRAV12-2. This bias is already present in A2/LLW-specific naïve T cells before vaccination with YF-17D. Using CD8+ T cell clones, we show that TRAV12-2 does not confer a functional advantage on a per cell basis. Molecular modeling indicated that the germline-encoded complementarity determining region (CDR) 1α loop of TRAV12-2 critically contributes to A2/LLW binding, in contrast to the conventional dominant dependence on somatically rearranged CDR3 loops. This germline component of antigen recognition may explain the unusually high precursor frequency, prevalence and immunodominance of T-cell responses specific for the A2/LLW epitope.
Collapse
Affiliation(s)
- Amandine Bovay
- Department of OncologyLausanne University Hospital (CHUV)EpalingesSwitzerland
| | - Vincent Zoete
- SIB Swiss Institute of BioinformaticsMolecular Modeling GroupLausanneSwitzerland
| | - Garry Dolton
- Division of Infection and Immunity and Systems Immunity Research InstituteCardiff University School of MedicineHeath ParkCardiffUK
| | - Anna M. Bulek
- Division of Infection and Immunity and Systems Immunity Research InstituteCardiff University School of MedicineHeath ParkCardiffUK
| | - David K. Cole
- Division of Infection and Immunity and Systems Immunity Research InstituteCardiff University School of MedicineHeath ParkCardiffUK
| | - Pierre J. Rizkallah
- Division of Infection and Immunity and Systems Immunity Research InstituteCardiff University School of MedicineHeath ParkCardiffUK
| | - Anna Fuller
- Division of Infection and Immunity and Systems Immunity Research InstituteCardiff University School of MedicineHeath ParkCardiffUK
| | - Konrad Beck
- Cardiff University School of DentistryHeath ParkCardiffUK
| | - Olivier Michielin
- SIB Swiss Institute of BioinformaticsMolecular Modeling GroupLausanneSwitzerland
| | - Daniel E. Speiser
- Department of OncologyLausanne University Hospital (CHUV)EpalingesSwitzerland
| | - Andrew K. Sewell
- Division of Infection and Immunity and Systems Immunity Research InstituteCardiff University School of MedicineHeath ParkCardiffUK
| | | |
Collapse
|
74
|
Jandus C, Usatorre AM, Viganò S, Zhang L, Romero P. The Vast Universe of T Cell Diversity: Subsets of Memory Cells and Their Differentiation. Methods Mol Biol 2018; 1514:1-17. [PMID: 27787788 DOI: 10.1007/978-1-4939-6548-9_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The T cell receptor confers specificity for antigen recognition to T cells. By the first encounter with the cognate antigen, reactive T cells initiate a program of expansion and differentiation that will define not only the ultimate quantity of specific cells that will be generated, but more importantly their quality and functional heterogeneity. Recent achievements using mouse model infection systems have helped to shed light into the complex network of factors that dictate and sustain memory T cell differentiation, ranging from antigen load, TCR signal strength, metabolic fitness, transcriptional programs, and proliferative potential. The different models of memory T cell differentiation are discussed in this chapter, and key phenotypic and functional attributes of memory T cell subsets are presented, both for mouse and human cells. Therapeutic manipulation of memory T cell generation is expected to provide novel unique ways to optimize current immunotherapies, both in infection and cancer.
Collapse
Affiliation(s)
- Camilla Jandus
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Amaia Martínez Usatorre
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Selena Viganò
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Lianjun Zhang
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Pedro Romero
- Translational Tumor Immunology Group, Ludwig Cancer Research Center, University of Lausanne, Biopole III, CB02, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| |
Collapse
|
75
|
CD28neg. T lymphocytes of a melanoma patient harbor tumor immunity and a high frequency of germline-encoded and public TCRs. Immunol Res 2017; 66:79-86. [DOI: 10.1007/s12026-017-8976-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
76
|
Butyrate and propionate inhibit antigen-specific CD8 + T cell activation by suppressing IL-12 production by antigen-presenting cells. Sci Rep 2017; 7:14516. [PMID: 29109552 PMCID: PMC5673935 DOI: 10.1038/s41598-017-15099-w] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/20/2017] [Indexed: 12/15/2022] Open
Abstract
Short chain fatty acids (SCFAs), such as acetate, butyrate and propionate, are products of microbial macronutrients fermentation that distribute systemically and are believed to modulate host immune responses. Recent data have indicated that certain SCFAs, such as butyrate and propionate, directly modulate human dendritic cell (DC) function. Given the role of DCs in initiating and shaping the adaptive immune response, we now explore how SCFAs affect the activation of antigen-specific CD8+ T cells stimulated with autologous, MART1 peptide-pulsed DC. We show that butyrate reduces the frequency of peptide-specific CD8+ T cells and, together with propionate, inhibit the activity of those cells. On the contrary, acetate does not affect them. Importantly, butyrate and propionate inhibit the production of IL-12 and IL-23 in the DCs and exogenous IL-12 fully restores the activation of the MART-1-specific CD8+ T cells, whereas IL-23 has no effect. In conclusion, these results point to a pivotal role of butyrate and propionate in modulating CD8+ T cell activation via the inhibition of IL-12 secretion from DCs. These findings reveal a novel mechanism whereby bacterial fermentation products may modulate CD8+ T cell function with possible implications in anti-cancer immunotherapy.
Collapse
|
77
|
Martin SD, Wick DA, Nielsen JS, Little N, Holt RA, Nelson BH. A library-based screening method identifies neoantigen-reactive T cells in peripheral blood prior to relapse of ovarian cancer. Oncoimmunology 2017; 7:e1371895. [PMID: 29296522 PMCID: PMC5739566 DOI: 10.1080/2162402x.2017.1371895] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/18/2017] [Accepted: 08/19/2017] [Indexed: 12/22/2022] Open
Abstract
Mutated cancer antigens, or neoantigens, represent compelling immunological targets and appear to underlie the success of several forms of immunotherapy. While there are anecdotal reports of neoantigen-specific T cells being present in the peripheral blood and/or tumors of cancer patients, effective adoptive cell therapy (ACT) against neoantigens will require reliable methods to isolate and expand rare, neoantigen-specific T cells from clinically available biospecimens, ideally prior to clinical relapse. Here, we addressed this need using “mini-lines”, large libraries of parallel T cell cultures, each originating from only 2,000 T cells. Using small quantities of peripheral blood from multiple time points in an ovarian cancer patient, we screened over 3.3 × 106 CD8+ T cells by ELISPOT for recognition of peptides corresponding to the full complement of somatic mutations (n = 37) from the patient's tumor. We identified ten T cell lines which collectively recognized peptides encoding five distinct mutations. Six of the ten T cell lines recognized a previously described neoantigen from this patient (HSDL1L25V), whereas the remaining four lines recognized peptides corresponding to four other mutations. Only the HSDL1L25V-specific T cell lines recognized autologous tumor. HSDL1L25V-specific T cells comprised at least three distinct clonotypes and could be identified and expanded from peripheral blood 3–9 months prior to the first tumor recurrence. These T cells became undetectable at later time points, underscoring the dynamic nature of the response. Thus, neoantigen-specific T cells can be expanded from small volumes of blood during tumor remission, making pre-emptive ACT a plausible clinical strategy.
Collapse
Affiliation(s)
- Spencer D Martin
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Michael Smith's Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Darin A Wick
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - Julie S Nielsen
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - Nicole Little
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Robert A Holt
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Michael Smith's Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Brad H Nelson
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
78
|
Bianchi F, Textor J, van den Bogaart G. Transmembrane Helices Are an Overlooked Source of Major Histocompatibility Complex Class I Epitopes. Front Immunol 2017; 8:1118. [PMID: 28959259 PMCID: PMC5604083 DOI: 10.3389/fimmu.2017.01118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/25/2017] [Indexed: 11/13/2022] Open
Abstract
About a fourth of the human proteome is anchored by transmembrane helices (TMHs) to lipid membranes. TMHs require multiple hydrophobic residues for spanning membranes, and this shows a striking resemblance with the requirements for peptide binding to major histocompatibility complex (MHC) class I. It, therefore, comes as no surprise that bioinformatics analysis predicts an over-representation of TMHs among strong MHC class I (MHC-I) binders. Published peptide elution studies confirm that TMHs are indeed presented by MHC-I. This raises the question how membrane proteins are processed for MHC-I (cross-)presentation, with current research focusing on soluble antigens. The presentation of membrane-buried peptides is likely important in health and disease, as TMHs are considerably conserved and their presentation might prevent escape mutations by pathogens. Therefore, it could contribute to the disease correlations described for many human leukocyte antigen haplotypes.
Collapse
Affiliation(s)
- Frans Bianchi
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
79
|
Graft versus self (GvS) against T-cell autoantigens is a mechanism of graft-host interaction. Proc Natl Acad Sci U S A 2016; 113:13827-13832. [PMID: 27834728 DOI: 10.1073/pnas.1609118113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Graft-versus-host disease (GVHD) represents the major nonrelapse complication of allogeneic hematopoietic cell transplantation. Although rare, the CNS and the eye can be affected. In this study, manifestation in the retina as part of the CNS and T-cell epitopes recognized by the allogeneic T cells were evaluated. In 2 of 6 patients with posttransplantation retina diseases and 6 of 22 patients without ocular symptoms, antigen-specific T-cell responses against retina-specific epitopes were observed. No genetic differences between donor and recipient could be identified indicating T-cell activation against self-antigens (graft versus self). Transplantation of a preexisting immunity and cross-reactivity with ubiquitous epitopes was excluded in family donors and healthy individuals. In summary, an immunological reaction against retina cells represents a mechanism of graft-versus-host interaction following hematopoietic cell transplantation.
Collapse
|
80
|
Abstract
Adoptive cellular therapy represents a robust means of augmenting the tumor-reactive effector population in patients with cancer by adoptive transfer of ex vivo expanded T cells. Three approaches have been developed to achieve this goal: the use of tumor-infiltrating lymphocytes or tumor-infiltrating lymphocytess extracted from patient biopsy material; the redirected engineering of lymphocytes using vectors expressing a chimeric antigen receptor and T-cell receptor; and third, the isolation and expansion of often low-frequency endogenous T cells (ETCs) reactive to tumor antigens from the peripheral blood of patients. This last form of adoptive transfer of T cells, known as ETC therapy, requires specialized methods to isolate and expand from peripheral blood the very low-frequency tumor-reactive T cells, methods that have been developed over the last 2 decades, to the point where such an approach may be broadly applicable not only for the treatment of melanoma but also for that of other solid tumor malignancies. One compelling feature of ETC is the ability to rapidly deploy clinical trials following identification of a tumor-associated target epitope, a feature that may be exploited to develop personalized antigen-specific T-cell therapy for patients with almost any solid tumor. With a well-validated antigen discovery pipeline in place, clinical studies combining ETC with agents that modulate the immune microenvironment can be developed that will transform ETC into a feasible treatment modality.
Collapse
|
81
|
Fuertes Marraco SA, Soneson C, Cagnon L, Gannon PO, Allard M, Abed Maillard S, Montandon N, Rufer N, Waldvogel S, Delorenzi M, Speiser DE. Long-lasting stem cell-like memory CD8+ T cells with a naïve-like profile upon yellow fever vaccination. Sci Transl Med 2016; 7:282ra48. [PMID: 25855494 DOI: 10.1126/scitranslmed.aaa3700] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Efficient and persisting immune memory is essential for long-term protection from infectious and malignant diseases. The yellow fever (YF) vaccine is a live attenuated virus that mediates lifelong protection, with recent studies showing that the CD8(+) T cell response is particularly robust. Yet, limited data exist regarding the long-term CD8(+) T cell response, with no studies beyond 5 years after vaccination. We investigated 41 vaccinees, spanning 0.27 to 35 years after vaccination. YF-specific CD8(+) T cells were readily detected in almost all donors (38 of 41), with frequencies decreasing with time. As previously described, effector cells dominated the response early after vaccination. We detected a population of naïve-like YF-specific CD8(+) T cells that was stably maintained for more than 25 years and was capable of self-renewal ex vivo. In-depth analyses of markers and genome-wide mRNA profiling showed that naïve-like YF-specific CD8(+) T cells in vaccinees (i) were distinct from genuine naïve cells in unvaccinated donors, (ii) resembled the recently described stem cell-like memory subset (Tscm), and (iii) among all differentiated subsets, had profiles closest to naïve cells. Our findings reveal that CD8(+) Tscm are efficiently induced by a vaccine in humans, persist for decades, and preserve a naïveness-like profile. These data support YF vaccination as an optimal mechanistic model for the study of long-lasting memory CD8(+) T cells in humans.
Collapse
Affiliation(s)
- Silvia A Fuertes Marraco
- Ludwig Cancer Center, University of Lausanne, Epalinges CH-1066, Switzerland. Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland
| | - Charlotte Soneson
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne CH-1015, Switzerland
| | - Laurène Cagnon
- Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland
| | - Philippe O Gannon
- Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland
| | - Mathilde Allard
- Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland
| | - Samia Abed Maillard
- Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland
| | - Nicole Montandon
- Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland
| | - Nathalie Rufer
- Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland
| | - Sophie Waldvogel
- Service Vaudois de Transfusion Sanguine de la Croix Rouge, Epalinges CH-1066, Switzerland
| | - Mauro Delorenzi
- Ludwig Cancer Center, University of Lausanne, Epalinges CH-1066, Switzerland. Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland. Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne CH-1015, Switzerland
| | - Daniel E Speiser
- Ludwig Cancer Center, University of Lausanne, Epalinges CH-1066, Switzerland. Department of Oncology, University Hospital of Lausanne (Centre Hospitalier Universitaire Vaudois), Lausanne CH-1011, Switzerland.
| |
Collapse
|
82
|
Lissina A, Briceño O, Afonso G, Larsen M, Gostick E, Price DA, Mallone R, Appay V. Priming of Qualitatively Superior Human Effector CD8+ T Cells Using TLR8 Ligand Combined with FLT3 Ligand. THE JOURNAL OF IMMUNOLOGY 2015; 196:256-263. [PMID: 26608912 DOI: 10.4049/jimmunol.1501140] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/08/2015] [Indexed: 01/01/2023]
Abstract
The quality of Ag-specific CD8(+) T cell responses is central to immune efficacy in infectious and malignant settings. Inducing effector CD8(+) T cells with potent functional properties is therefore a priority in the field of immunotherapy. However, the optimal assessment of new treatment strategies in humans is limited by currently available testing platforms. In this study, we introduce an original model of in vitro CD8(+) T cell priming, based on an accelerated dendritic cell coculture system, which uses unfractionated human PBMCs as the starting material. This approach enables the rapid evaluation of adjuvant effects on the functional properties of human CD8(+) T cells primed from Ag-specific naive precursors. We demonstrate that a selective TLR8 agonist, in combination with FLT3L, primes high-quality CD8(+) T cell responses. TLR8L/FLT3L-primed CD8(+) T cells displayed enhanced cytotoxic activity, polyfunctionality, and Ag sensitivity. The acquisition of this superior functional profile was associated with increased T-bet expression induced via an IL-12-dependent mechanism. Collectively, these data validate an expedited route to vaccine delivery or optimal T cell expansion for adoptive cell transfer.
Collapse
Affiliation(s)
- Anna Lissina
- Sorbonne Universités, UPMC Univ Paris 06, DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,INSERM, U1135, CIMI-Paris, Paris, France
| | - Olivia Briceño
- Sorbonne Universités, UPMC Univ Paris 06, DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,INSERM, U1135, CIMI-Paris, Paris, France
| | - Georgia Afonso
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Martin Larsen
- Sorbonne Universités, UPMC Univ Paris 06, DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,INSERM, U1135, CIMI-Paris, Paris, France
| | - Emma Gostick
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, Wales, UK
| | - David A Price
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, Wales, UK
| | - Roberto Mallone
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Institut Cochin, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service de Diabétologie, Paris, France
| | - Victor Appay
- Sorbonne Universités, UPMC Univ Paris 06, DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,INSERM, U1135, CIMI-Paris, Paris, France
| |
Collapse
|
83
|
STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc Natl Acad Sci U S A 2015; 112:15408-13. [PMID: 26607445 DOI: 10.1073/pnas.1512832112] [Citation(s) in RCA: 437] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Spontaneous CD8 T-cell responses occur in growing tumors but are usually poorly effective. Understanding the molecular and cellular mechanisms that drive these responses is of major interest as they could be exploited to generate a more efficacious antitumor immunity. As such, stimulator of IFN genes (STING), an adaptor molecule involved in cytosolic DNA sensing, is required for the induction of antitumor CD8 T responses in mouse models of cancer. Here, we find that enforced activation of STING by intratumoral injection of cyclic dinucleotide GMP-AMP (cGAMP), potently enhanced antitumor CD8 T responses leading to growth control of injected and contralateral tumors in mouse models of melanoma and colon cancer. The ability of cGAMP to trigger antitumor immunity was further enhanced by the blockade of both PD1 and CTLA4. The STING-dependent antitumor immunity, either induced spontaneously in growing tumors or induced by intratumoral cGAMP injection was dependent on type I IFNs produced in the tumor microenvironment. In response to cGAMP injection, both in the mouse melanoma model and an ex vivo model of cultured human melanoma explants, the principal source of type I IFN was not dendritic cells, but instead endothelial cells. Similarly, endothelial cells but not dendritic cells were found to be the principal source of spontaneously induced type I IFNs in growing tumors. These data identify an unexpected role of the tumor vasculature in the initiation of CD8 T-cell antitumor immunity and demonstrate that tumor endothelial cells can be targeted for immunotherapy of melanoma.
Collapse
|
84
|
Alanio C, Nicoli F, Sultanik P, Flecken T, Perot B, Duffy D, Bianchi E, Lim A, Clave E, van Buuren MM, Schnuriger A, Johnsson K, Boussier J, Garbarg-Chenon A, Bousquet L, Mottez E, Schumacher TN, Toubert A, Appay V, Heshmati F, Thimme R, Pol S, Mallet V, Albert ML. Bystander hyperactivation of preimmune CD8+ T cells in chronic HCV patients. eLife 2015; 4. [PMID: 26568315 PMCID: PMC4752008 DOI: 10.7554/elife.07916] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 11/12/2015] [Indexed: 01/21/2023] Open
Abstract
Chronic infection perturbs immune homeostasis. While prior studies have reported dysregulation of effector and memory cells, little is known about the effects on naïve T cell populations. We performed a cross-sectional study of chronic hepatitis C (cHCV) patients using tetramer-associated magnetic enrichment to study antigen-specific inexperienced CD8(+) T cells (i.e., tumor or unrelated virus-specific populations in tumor-free and sero-negative individuals). cHCV showed normal precursor frequencies, but increased proportions of memory-phenotype inexperienced cells, as compared to healthy donors or cured HCV patients. These observations could be explained by low surface expression of CD5, a negative regulator of TCR signaling. Accordingly, we demonstrated TCR hyperactivation and generation of potent CD8(+) T cell responses from the altered T cell repertoire of cHCV patients. In sum, we provide the first evidence that naïve CD8(+) T cells are dysregulated during cHCV infection, and establish a new mechanism of immune perturbation secondary to chronic infection.
Collapse
Affiliation(s)
- Cécile Alanio
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Francesco Nicoli
- Sorbonne Universités, UPMC Univ Paris 06, DNU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,Emory, , United States
| | - Philippe Sultanik
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Tobias Flecken
- The University Medical Center Freiburg, Department of Internal Medicine II, Albert-Ludwigs-Universität, Freiberg, Germany
| | - Brieuc Perot
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Darragh Duffy
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | | | - Annick Lim
- Plateforme d'Immunoscope, Institut Pasteur, Paris, France
| | - Emmanuel Clave
- Hôpital Saint-Louis, Assistance publique - hôpitaux de Paris, Paris, France
| | - Marit M van Buuren
- Department of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Aurélie Schnuriger
- Laboratoire de virologie, Hôpital Armand-Trousseau, Assistance publique - hôpitaux de Paris, Paris, France
| | - Kerstin Johnsson
- Mathematics, Faculty of Engineering, Lunds University, Lund, Sweden
| | - Jeremy Boussier
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| | - Antoine Garbarg-Chenon
- Laboratoire de virologie, Hôpital Armand-Trousseau, Assistance publique - hôpitaux de Paris, Paris, France
| | | | - Estelle Mottez
- Centre d'Immunologie Humaine, Institut Pasteur, Paris, France
| | - Ton N Schumacher
- Department of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Antoine Toubert
- Hôpital Saint-Louis, Assistance publique - hôpitaux de Paris, Paris, France
| | - Victor Appay
- Sorbonne Universités, UPMC Univ Paris 06, DNU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,Emory, , United States
| | | | - Robert Thimme
- The University Medical Center Freiburg, Department of Internal Medicine II, Albert-Ludwigs-Universität, Freiberg, Germany
| | | | | | - Matthew L Albert
- Unités de Recherche Internationales Mixtes Pasteur, Institut Pasteur, Paris, France.,Centre d'Immunologie Humaine, Institut Pasteur, Paris, France.,Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
| |
Collapse
|
85
|
Thaxton JE, Li Z. To affinity and beyond: harnessing the T cell receptor for cancer immunotherapy. Hum Vaccin Immunother 2015; 10:3313-21. [PMID: 25483644 DOI: 10.4161/21645515.2014.973314] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
T cell adoptive therapies for immune-mediated regression of cancers have attracted a great deal of recent attention. Clinical results are glamorous, yet much remains to be uncovered behind the basic science that allows us to engineer T cells and T cell receptors (TCRs) for clinical use. We discuss the development of TCRs for therapeutic use in the context of thymic selection toward central tolerance and we review therapies based on tumor infiltrating lymphocytes (TILs), endogenous antigen specific TCRs, and engineered TCRs. Further we discuss the development of low and high affinity TCRs and the extent to which each challenges central tolerance. Current results suggest that adaptation of TCR engineering of moderate affinity TCRs coupled with co-regulatory and stimulatory molecules may be the safest and most efficacious road for TCR development aimed at tumor abolition.
Collapse
Key Words
- AIRE, autoimmune regulator
- CDR, complementarity determining region
- CTA, cancer testis antigen
- MHC, major histocompatibility complex
- SLEC, short-lived effector cell
- T cell receptor
- TAA, tumor-associated antigen
- TCR, T cell receptor
- TIL, tumor infiltrating lymphocyte
- TSA, tissue-specific self-antigen
- adoptive cell therapy
- affinity
- cancer
- co-receptor
- mTEC, medullary thymic epithelial cell
- tumor
Collapse
Affiliation(s)
- Jessica E Thaxton
- a Department of Microbiology and Immunology; Hollings Cancer Center ; Medical University of South Carolina ; Charleston , SC USA
| | | |
Collapse
|
86
|
Tscharke DC, Croft NP, Doherty PC, La Gruta NL. Sizing up the key determinants of the CD8+ T cell response. Nat Rev Immunol 2015; 15:705-16. [DOI: 10.1038/nri3905] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
87
|
Wang M, Yin B, Wang HY, Wang RF. Current advances in T-cell-based cancer immunotherapy. Immunotherapy 2015; 6:1265-78. [PMID: 25524383 DOI: 10.2217/imt.14.86] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer is a leading cause of death worldwide; due to the lack of ideal cancer biomarkers for early detection or diagnosis, most patients present with late-stage disease at the time of diagnosis, thus limiting the potential for successful treatment. Traditional cancer treatments, including surgery, chemotherapy and radiation therapy, have demonstrated very limited efficacy for patients with late-stage disease. Therefore, innovative and effective cancer treatments are urgently needed for cancer patients with late-stage and refractory disease. Cancer immunotherapy, particularly adoptive cell transfer, has shown great promise in the treatment of patients with late-stage disease, including those who are refractory to standard therapies. In this review, we will highlight recent advances and discuss future directions in adoptive cell transfer based cancer immunotherapy.
Collapse
Affiliation(s)
- Mingjun Wang
- Center for Inflammation & Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
88
|
Yu W, Jiang N, Ebert PJR, Kidd BA, Müller S, Lund PJ, Juang J, Adachi K, Tse T, Birnbaum ME, Newell EW, Wilson DM, Grotenbreg GM, Valitutti S, Quake SR, Davis MM. Clonal Deletion Prunes but Does Not Eliminate Self-Specific αβ CD8(+) T Lymphocytes. Immunity 2015; 42:929-41. [PMID: 25992863 DOI: 10.1016/j.immuni.2015.05.001] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 02/27/2015] [Accepted: 03/23/2015] [Indexed: 02/06/2023]
Abstract
It has long been thought that clonal deletion efficiently removes almost all self-specific T cells from the peripheral repertoire. We found that self-peptide MHC-specific CD8(+) T cells in the blood of healthy humans were present in frequencies similar to those specific for non-self antigens. For the Y chromosome-encoded SMCY antigen, self-specific T cells exhibited only a 3-fold lower average frequency in males versus females and were anergic with respect to peptide activation, although this inhibition could be overcome by a stronger stimulus. We conclude that clonal deletion prunes but does not eliminate self-specific T cells and suggest that to do so would create holes in the repertoire that pathogens could readily exploit. In support of this hypothesis, we detected T cells specific for all 20 amino acid variants at the p5 position of a hepatitis C virus epitope in a random group of blood donors.
Collapse
Affiliation(s)
- Wong Yu
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ning Jiang
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter J R Ebert
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian A Kidd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sabina Müller
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France and Université Toulouse III Paul-Sabatier, 31024 Toulouse, France
| | - Peder J Lund
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeremy Juang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Keishi Adachi
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tiffany Tse
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael E Birnbaum
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Evan W Newell
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Darrell M Wilson
- Department of Pediatric Endocrinology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Salvatore Valitutti
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France and Université Toulouse III Paul-Sabatier, 31024 Toulouse, France
| | - Stephen R Quake
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
89
|
Schmied S, Gostick E, Price DA, Abken H, Assenmacher M, Richter A. Analysis of the functional WT1-specific T-cell repertoire in healthy donors reveals a discrepancy between CD4(+) and CD8(+) memory formation. Immunology 2015; 145:558-69. [PMID: 25882672 PMCID: PMC4515135 DOI: 10.1111/imm.12472] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/11/2015] [Accepted: 03/31/2015] [Indexed: 01/03/2023] Open
Abstract
The Wilms' tumour-1 (WT1) protein is considered a prime target for cancer immunotherapy based on its presumptive immunogenicity and widespread expression across a variety of malignancies. However, little is known about the naturally occurring WT1-specific T-cell repertoire because self-derived antigens typically elicit low frequency responses that challenge the sensitivity limits of current detection techniques. In this study, we used highly efficient cell enrichment procedures based on CD137, CD154, and pHLA class I tetramer staining to conduct a detailed analysis of WT1-specific T cells from the peripheral blood. Remarkably, we detected WT1-specific CD4(+) and CD8(+) T-cell populations in the vast majority of healthy individuals. Memory responses specific for WT1 were commonly present in the CD4(+) T-cell compartment, whereas WT1-specific CD8(+) T cells almost universally displayed a naive phenotype. Moreover, memory CD4(+) and naive CD8(+) T cells with specificity for WT1 were found to coexist in some individuals. Collectively, these findings suggest a natural discrepancy between the CD4(+) and CD8(+) T-cell lineages with respect to memory formation in response to a self-derived antigen. Nonetheless, WT1-specific T cells from both lineages were readily activated ex vivo and expanded in vitro, supporting the use of strategies designed to exploit this expansive reservoir of self-reactive T cells for immunotherapeutic purposes.
Collapse
Affiliation(s)
| | - Emma Gostick
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - David A Price
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Hinrich Abken
- Centre for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Department I Internal Medicine, University Hospital Cologne, Cologne, Germany
| | | | - Anne Richter
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| |
Collapse
|
90
|
Human Tumor Antigens and Cancer Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:948501. [PMID: 26161423 PMCID: PMC4487697 DOI: 10.1155/2015/948501] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/03/2015] [Indexed: 01/21/2023]
Abstract
With the recent developments of adoptive T cell therapies and the use of new monoclonal antibodies against the immune checkpoints, immunotherapy is at a turning point. Key players for the success of these therapies are the cytolytic T lymphocytes, which are a subset of T cells able to recognize and kill tumor cells. Here, I review the nature of the antigenic peptides recognized by these T cells and the processes involved in their presentation. I discuss the importance of understanding how each antigenic peptide is processed in the context of immunotherapy and vaccine delivery.
Collapse
|
91
|
Kissick HT, Sanda MG. The role of active vaccination in cancer immunotherapy: lessons from clinical trials. Curr Opin Immunol 2015; 35:15-22. [PMID: 26050634 DOI: 10.1016/j.coi.2015.05.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 01/05/2023]
Abstract
In the past few years, a number of different immunotherapeutic strategies have shown impressive results in cancer patients. These successful approaches include blockade of immunosuppressive molecules like PD-1 and CTLA-4, adoptive transfer of patient derived and genetically modified T-cells, and vaccines that stimulate tumor antigen specific T-cells. However, several large vaccine trials recently failed to reach designated primary endpoints. In light of the success of other immunotherapeutic approaches, these negative results raise the questions of why vaccines have not generated a better response, and what the role of active vaccination will be moving forward in cancer immunotherapy.
Collapse
Affiliation(s)
- Haydn T Kissick
- Department of Urology, Emory University School of Medicine, United States; Department of Microbiology and Immunology, Emory University School of Medicine, United States.
| | - Martin G Sanda
- Department of Urology, Emory University School of Medicine, United States
| |
Collapse
|
92
|
Klaver Y, Kunert A, Sleijfer S, Debets R, Lamers CHJ. Adoptive T-cell therapy: a need for standard immune monitoring. Immunotherapy 2015; 7:513-33. [DOI: 10.2217/imt.15.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer immune therapy, in particular the use of checkpoint inhibitors and adoptive transfer of T cells has recently demonstrated significant clinical responses against several tumor types. Unfortunately, these therapies are frequently accompanied by severe toxicities, underscoring the need for markers that provide information on therapy response. Monitoring immune responses in the tumor microenvironment and peripheral blood prior to and during these therapies will provide better insight into the mechanisms underlying clinical activities, and will potentially enable the identification of such markers. In this review, we present an overview of adoptive T-cell trials conducted with a special focus on immune monitoring, and argue that accurate monitoring of T cells is pivotal to further development of immune therapies to treat cancer.
Collapse
Affiliation(s)
- Yarne Klaver
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Cor HJ Lamers
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
93
|
Chevalier MF, Bobisse S, Costa-Nunes C, Cesson V, Jichlinski P, Speiser DE, Harari A, Coukos G, Romero P, Nardelli-Haefliger D, Jandus C, Derré L. High-throughput monitoring of human tumor-specific T-cell responses with large peptide pools. Oncoimmunology 2015; 4:e1029702. [PMID: 26451296 DOI: 10.1080/2162402x.2015.1029702] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/06/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022] Open
Abstract
In immune intervention trials, the comprehensive investigation of immunogenicity or T-cell epitope-mapping is challenging especially when a large set of epitopes needs to be screened and limited sample material is available. To this end, T-cell responses are often monitored using peptide pools. Here, we assessed the magnitude and sensitivity of detection of antigen-specific CD8+ and CD4+ T cells using a single peptide alone or mixed into large pools. Interestingly the magnitude of ex vivo anti-viral and anti-tumor T-cell responses was identical irrespective of the presence and number of irrelevant peptides, in different functional assays with PBMCs from healthy donors and cancer patients. Moreover, the presence of up to 300 irrelevant peptides did not affect the threshold of responsiveness of antigen-specific CD8+ T cells to single cognate peptides. These data demonstrate the relevance of using very large peptide pools for the sensitive and specific immune-monitoring of epitope-specific T cells in natural or immune-modulated context.
Collapse
Affiliation(s)
- Mathieu F Chevalier
- Urology Research Unit; Department of Urology; University Hospital of Lausanne (CHUV) ; Lausanne, Switzerland
| | - Sara Bobisse
- Center of Experimental Therapeutics; Department of Oncology; University Hospital of Lausanne ; Lausanne, Switzerland ; Ludwig Center for Cancer Research at University of Lausanne ; Epalinges, Switzerland
| | - Carla Costa-Nunes
- Ludwig Center for Cancer Research at University of Lausanne ; Epalinges, Switzerland
| | - Valérie Cesson
- Urology Research Unit; Department of Urology; University Hospital of Lausanne (CHUV) ; Lausanne, Switzerland
| | - Patrice Jichlinski
- Urology Research Unit; Department of Urology; University Hospital of Lausanne (CHUV) ; Lausanne, Switzerland
| | - Daniel E Speiser
- Ludwig Center for Cancer Research at University of Lausanne ; Epalinges, Switzerland
| | - Alexandre Harari
- Center of Experimental Therapeutics; Department of Oncology; University Hospital of Lausanne ; Lausanne, Switzerland ; Ludwig Center for Cancer Research at University of Lausanne ; Epalinges, Switzerland
| | - George Coukos
- Ludwig Center for Cancer Research at University of Lausanne ; Epalinges, Switzerland ; Ovarian Cancer Research Center; Perelman School of Medicine; University of Pennsylvania ; Philadelphia, PA USA ; Department of Oncology; University Hospital of Lausanne (CHUV) ; Lausanne, Switzerland
| | - Pedro Romero
- Ludwig Center for Cancer Research at University of Lausanne ; Epalinges, Switzerland
| | - Denise Nardelli-Haefliger
- Urology Research Unit; Department of Urology; University Hospital of Lausanne (CHUV) ; Lausanne, Switzerland
| | - Camilla Jandus
- Ludwig Center for Cancer Research at University of Lausanne ; Epalinges, Switzerland
| | - Laurent Derré
- Urology Research Unit; Department of Urology; University Hospital of Lausanne (CHUV) ; Lausanne, Switzerland
| |
Collapse
|
94
|
Roberto A, Castagna L, Zanon V, Bramanti S, Crocchiolo R, McLaren JE, Gandolfi S, Tentorio P, Sarina B, Timofeeva I, Santoro A, Carlo-Stella C, Bruno B, Carniti C, Corradini P, Gostick E, Ladell K, Price DA, Roederer M, Mavilio D, Lugli E. Role of naive-derived T memory stem cells in T-cell reconstitution following allogeneic transplantation. Blood 2015; 125:2855-64. [PMID: 25742699 PMCID: PMC4424633 DOI: 10.1182/blood-2014-11-608406] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/28/2015] [Indexed: 12/31/2022] Open
Abstract
Early T-cell reconstitution following allogeneic transplantation depends on the persistence and function of T cells that are adoptively transferred with the graft. Posttransplant cyclophosphamide (pt-Cy) effectively prevents alloreactive responses from unmanipulated grafts, but its effect on subsequent immune reconstitution remains undetermined. Here, we show that T memory stem cells (TSCM), which demonstrated superior reconstitution capacity in preclinical models, are the most abundant circulating T-cell population in the early days following haploidentical transplantation combined with pt-Cy and precede the expansion of effector cells. Transferred naive, but not TSCM or conventional memory cells preferentially survive cyclophosphamide, thus suggesting that posttransplant TSCM originate from naive precursors. Moreover, donor naive T cells specific for exogenous and self/tumor antigens persist in the host and contribute to peripheral reconstitution by differentiating into effectors. Similarly, pathogen-specific memory T cells generate detectable recall responses, but only in the presence of the cognate antigen. We thus define the cellular basis of T-cell reconstitution following pt-Cy at the antigen-specific level and propose to explore naive-derived TSCM in the clinical setting to overcome immunodeficiency. These trials were registered at www.clinicaltrials.gov as #NCT02049424 and #NCT02049580.
Collapse
Affiliation(s)
- Alessandra Roberto
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Luca Castagna
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Veronica Zanon
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Stefania Bramanti
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Roberto Crocchiolo
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - James E McLaren
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Sara Gandolfi
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Paolo Tentorio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Barbara Sarina
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Inna Timofeeva
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Armando Santoro
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy
| | - Carmelo Carlo-Stella
- Hematology and Bone Marrow Transplant Unit, Humanitas Cancer Center, Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Cristiana Carniti
- Department of Haematology and Pediatric Onco-Haematology, Istituto Nazionale Tumori, Milan, Italy; and
| | - Paolo Corradini
- Department of Haematology and Pediatric Onco-Haematology, Istituto Nazionale Tumori, Milan, Italy; and
| | - Emma Gostick
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Kristin Ladell
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David A Price
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| |
Collapse
|
95
|
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
|
96
|
Zelba H, Weide B, Martens A, Bailur JK, Garbe C, Pawelec G. The prognostic impact of specific CD4 T-cell responses is critically dependent on the target antigen in melanoma. Oncoimmunology 2015; 4:e955683. [PMID: 26097797 DOI: 10.4161/21624011.2014.955683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/01/2014] [Indexed: 12/11/2022] Open
Abstract
The melanoma-associated antigens Melan-A and NY-ESO-1 stimulate different T-cell responses in late-stage melanoma patients. Either CD4+ or CD8+ T-cell reactivity against NY-ESO-1 was associated with better prognosis, but for Melan-A, only CD8+ but not CD4+ T-cell responses were associated with longer survival.
Collapse
Affiliation(s)
- Henning Zelba
- Interfaculty Institute for Cell Biology; Department of Immunology; University of Tübingen ; Tübingen, Germany
| | - Benjamin Weide
- Division of Dermatooncology; Department of Dermatology; Tübingen University Medical Center ; Tübingen, Germany
| | - Alexander Martens
- Department of Internal Medicine II; Section for Transplantation Immunology and Immunohematology; Tübingen University Medical Center ; Tübingen, Germany
| | - Jithendra Kini Bailur
- Department of Internal Medicine II; Section for Transplantation Immunology and Immunohematology; Tübingen University Medical Center ; Tübingen, Germany
| | - Claus Garbe
- Division of Dermatooncology; Department of Dermatology; Tübingen University Medical Center ; Tübingen, Germany
| | - Graham Pawelec
- Department of Internal Medicine II; Section for Transplantation Immunology and Immunohematology; Tübingen University Medical Center ; Tübingen, Germany
| |
Collapse
|
97
|
Fichtner S, Hose D, Engelhardt M, Meißner T, Neuber B, Krasniqi F, Raab M, Schönland S, Ho AD, Goldschmidt H, Hundemer M. Association of Antigen-Specific T-cell Responses with Antigen Expression and Immunoparalysis in Multiple Myeloma. Clin Cancer Res 2015; 21:1712-21. [DOI: 10.1158/1078-0432.ccr-14-1618] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 12/31/2014] [Indexed: 11/16/2022]
|
98
|
Maeda Y, Nishikawa H, Sugiyama D, Ha D, Hamaguchi M, Saito T, Nishioka M, Wing JB, Adeegbe D, Katayama I, Sakaguchi S. Detection of self-reactive CD8⁺ T cells with an anergic phenotype in healthy individuals. Science 2015; 346:1536-40. [PMID: 25525252 DOI: 10.1126/science.aaa1292] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Immunological tolerance to self requires naturally occurring regulatory T (Treg) cells. Yet how they stably control autoimmune T cells remains obscure. Here, we show that Treg cells can render self-reactive human CD8(+) T cells anergic (i.e., hypoproliferative and cytokine hypoproducing upon antigen restimulation) in vitro, likely by controlling the costimulatory function of antigen-presenting cells. Anergic T cells were naïve in phenotype, lower than activated T cells in T cell receptor affinity for cognate antigen, and expressed several coinhibitory molecules, including cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). Using these criteria, we detected in healthy individuals anergic T cells reactive with a skin antigen targeted in the autoimmune disease vitiligo. Collectively, our results suggest that Treg cell-mediated induction of anergy in autoimmune T cells is important for maintaining self-tolerance.
Collapse
Affiliation(s)
- Yuka Maeda
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan
| | - Hiroyoshi Nishikawa
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan.
| | - Daisuke Sugiyama
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan
| | - Danbee Ha
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan
| | - Masahide Hamaguchi
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan
| | - Takuro Saito
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan
| | - Megumi Nishioka
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan. Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - James B Wing
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan
| | - Dennis Adeegbe
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan
| | - Ichiro Katayama
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Shimon Sakaguchi
- Experimental Immunology, Immunology Frontier Research Center (IFReC-WPI), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
99
|
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: 4.1] [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
|
100
|
Romero P, Speiser DE, Rufer N. Deciphering the unusual HLA-A2/Melan-A/MART-1-specific TCR repertoire in humans. Eur J Immunol 2014; 44:2567-70. [PMID: 25154881 DOI: 10.1002/eji.201445004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 07/30/2014] [Accepted: 08/20/2014] [Indexed: 11/11/2022]
Abstract
The Melan-A/MART-1(26-35) antigenic peptide is one of the best studied human tumor-associated antigens. It is expressed in healthy melanocytes and malignant melanoma and is recognized by CD8(+) T cells in the context of the MHC class I molecule HLA-A*0201. While an unusually large repertoire of CD8(+) T cells specific for this antigen has been documented, the reasons for its generation have remained elusive. In this issue of the European Journal of Immunology, Pinto et al. [Eur. J. Immunol. 2014. 44: 2811-2821] uncover one important mechanism by comparing the thymic expression of the Melan-A gene to that in the melanocyte lineage. This study shows that medullary thymic epithelial cells (mTECs) dominantly express a truncated Melan-A transcript, the product of misinitiation of transcription. Consequently, the protein product in mTECs lacks the immunodominant epitope spanning residues 26-35, thus precluding central tolerance to this antigen. In contrast, melanocytes and melanoma tumor cells express almost exclusively the full-length Melan-A transcript, thus providing the target antigen for efficient recognition by HLA-A2-restricted CD8(+) T cells. The frequency of these alternative gene transcription modes may be more common than previously appreciated and may represent an important factor modulating the efficiency of central tolerance induction in the thymus.
Collapse
Affiliation(s)
- Pedro Romero
- Ludwig Cancer Research Center, Department of Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|