1
|
Matous JG, Snook JP, Contreras NA, Ramstead AG, Charley KR, Kolawole EM, Kisiolek JN, Flint KA, Soedel AJ, Robinson B, Mendoza AB, Kumaki Y, Evavold BD, Williams MA. Shp-1 regulates the activity of low-affinity T cells specific to endogenous self-antigen during melanoma tumor growth and drives resistance to immune checkpoint inhibition. J Immunother Cancer 2025; 13:e010879. [PMID: 40246583 PMCID: PMC12007028 DOI: 10.1136/jitc-2024-010879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND The presence of activated CD8 T cells in the tumor microenvironment is correlated with an effective immune response to immune checkpoint inhibitor (ICI) therapy. However, ICI predominantly targets high-affinity T cells, which may be less abundant in tumors with few neoantigens. Targeting the intracellular phosphatase Src homology region 2 domain-containing phosphatase-1 (Shp-1) in combination with ICI lowers the T cell activation threshold and enhances the ability of low-affinity T cells to mount a productive antitumor response. METHODS In this study, we sought to determine whether temporal inhibition of Shp-1 during active tumor growth could rescue the activity of low-affinity T cells specific for endogenous self-antigens. To address this question, we implanted Yale University Mouse Melanoma (YUMM) tumor cell lines into WT mice and, on tumor establishment, administered an inhibitor of Shp-1 (TPI-1) with or without ICI treatment. We analyzed treatment-dependent changes in the immune infiltrate in the tumor via flow cytometry, major histocompatibility complex (MHC) tetramer-mediated detection of tyrosinase-related protein 2 (TRP-2)180-188-specific T cells and a micropipette-based two-dimensional affinity assay to measure the T cell receptor (TCR) affinity. RESULTS Administration of ICI and a Shp-1 inhibitor to mice with established YUMM tumors, but neither agent alone, resulted in a significant delay in tumor growth and an increased frequency of CD8 tumor-infiltrating T cells with enhanced effector and reduced exhaustion characteristics. In particular, combined treatment increased the frequency of CD8 T cells specific for the MHC Class I-restricted tumor self-antigen TRP-2180-188. We found that the increase in effector T cells was almost entirely due to an increase in T cells with very low TCR affinity. CONCLUSIONS We conclude that approaches for altering TCR signaling threshold are effective in enhancing the antitumor response of low-affinity T cells specific for endogenous self-antigens in settings of ICI resistance and/or where neoantigens are not available to drive antitumor responses.
Collapse
Affiliation(s)
- Joseph G Matous
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Jeremy P Snook
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Nico A Contreras
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Andrew G Ramstead
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Krystal R Charley
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | | | - Jacob N Kisiolek
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
| | - Kaitlyn A Flint
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Ashleigh J Soedel
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Breyana Robinson
- North Carolina Agricultural and Technical State University, Greensboro, North Carolina, USA
| | | | - Yohichi Kumaki
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Brian D Evavold
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
| | - Matthew A Williams
- Department of Pathology, The University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| |
Collapse
|
2
|
Lopez de Rodas M, Villalba-Esparza M, Sanmamed MF, Chen L, Rimm DL, Schalper KA. Biological and clinical significance of tumour-infiltrating lymphocytes in the era of immunotherapy: a multidimensional approach. Nat Rev Clin Oncol 2025; 22:163-181. [PMID: 39820025 DOI: 10.1038/s41571-024-00984-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have improved clinical outcomes across several solid tumour types. Prominent efforts have focused on understanding the anticancer mechanisms of these agents, identifying biomarkers of response and uncovering resistance mechanisms to develop new immunotherapeutic approaches. This research has underscored the crucial roles of the tumour microenvironment and, particularly, tumour-infiltrating lymphocytes (TILs) in immune-mediated tumour elimination. Numerous studies have evaluated the prognostic and predictive value of TILs and the mechanisms that govern T cell dysfunction, fuelled by technical developments in single-cell transcriptomics, proteomics, high-dimensional spatial platforms and advanced computational models. However, questions remain regarding the definition of TILs, optimal strategies to study them, specific roles of different TIL subpopulations and their clinical implications in different treatment contexts. Additionally, most studies have focused on the abundance of major TIL subpopulations but have not developed standardized quantification strategies or analysed other crucial aspects such as their functional profile, spatial distribution and/or arrangement, tumour antigen-reactivity, clonal diversity and heterogeneity. In this Review, we discuss a conceptual framework for the systematic study of TILs and summarize the evidence regarding their biological properties and biomarker potential for ICI therapy. We also highlight opportunities, challenges and strategies to support future developments in this field.
Collapse
Affiliation(s)
- Miguel Lopez de Rodas
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba-Esparza
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada and Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Long J, Wang Y, Jiang X, Ge J, Chen M, Zheng B, Wang R, Wang M, Xu M, Ke Q, Wang J. Nanomaterials Boost CAR-T Therapy for Solid Tumors. Adv Healthc Mater 2024; 13:e2304615. [PMID: 38483400 DOI: 10.1002/adhm.202304615] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/29/2024] [Indexed: 05/22/2024]
Abstract
T cell engineering, particularly via chimeric antigen receptor (CAR) modifications for enhancing tumor specificity, has shown efficacy in treating hematologic malignancies. The extension of CAR-T cell therapy to solid tumors, however, is impeded by several challenges: The absence of tumor-specific antigens, antigen heterogeneity, a complex immunosuppressive tumor microenvironment, and physical barriers to cell infiltration. Additionally, limitations in CAR-T cell manufacturing capacity and the high costs associated with these therapies restrict their widespread application. The integration of nanomaterials into CAR-T cell production and application offers a promising avenue to mitigate these challenges. Utilizing nanomaterials in the production of CAR-T cells can decrease product variability and lower production expenses, positively impacting the targeting and persistence of CAR-T cells in treatment and minimizing adverse effects. This review comprehensively evaluates the use of various nanomaterials in the production of CAR-T cells, genetic modification, and in vivo delivery. It discusses their underlying mechanisms and potential for clinical application, with a focus on improving specificity and safety in CAR-T cell therapy.
Collapse
Affiliation(s)
- Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, 1001 Xueyuan Road, Shenzhen, 518055, China
| | - Yian Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, 410013, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Junshang Ge
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, 410078, China
| | - Mingfen Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Fujian Medical University, Quanzhou, 362000, China
| | - Boshu Zheng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Rong Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Meifang Xu
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Qi Ke
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| | - Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, No.1 Xuefu North Road University Town, Fuzhou, 350122, China
| |
Collapse
|
4
|
Laletin V, Bernard PL, Montersino C, Yamanashi Y, Olive D, Castellano R, Guittard G, Nunès JA. DOK1 and DOK2 regulate CD8 T cell signaling and memory formation without affecting tumor cell killing. Sci Rep 2024; 14:15053. [PMID: 38956389 PMCID: PMC11220026 DOI: 10.1038/s41598-024-66075-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Targeting intracellular inhibiting proteins has been revealed to be a promising strategy to improve CD8+ T cell anti-tumor efficacy. Here, we are focusing on intracellular inhibiting proteins specific to TCR signaling: DOK1 and DOK2 expressed in T cells. We hypothesized that depletion of intracellular inhibition checkpoint DOK1 and DOK2 could improve CD8+ T-cell based cancer therapies. To evaluate the role of DOK1 and DOK2 depletion in physiology and effector function of CD8+ T lymphocytes and in cancer progression, we established a transgenic T cell receptor mouse model specific to melanoma antigen hgp100 (pmel-1 TCR Tg) in WT and Dok1/Dok2 DKO (double KO) mice. We showed that both DOK1 and DOK2 depletion in CD8+ T cells after an in vitro pre-stimulation induced a higher percentage of effector memory T cells as well as an up regulation of TCR signaling cascade- induced by CD3 mAbs, including the increased levels of pAKT and pERK, two major phosphoproteins involved in T cell functions. Interestingly, this improved TCR signaling was not observed in naïve CD8+ T cells. Despite this enhanced TCR signaling essentially shown upon stimulation via CD3 mAbs, pre-stimulated Dok1/Dok2 DKO CD8+ T cells did not show any increase in their activation or cytotoxic capacities against melanoma cell line expressing hgp100 in vitro. Altogether we demonstrate here a novel aspect of the negative regulation by DOK1 and DOK2 proteins in CD8+ T cells. Indeed, our results allow us to conclude that DOK1 and DOK2 have an inhibitory role following long term T cell stimulations.
Collapse
Affiliation(s)
- Vladimir Laletin
- Centre de Recherche en Cancérologie de Marseille, CRCM, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Pierre-Louis Bernard
- Centre de Recherche en Cancérologie de Marseille, CRCM, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Camille Montersino
- Centre de Recherche en Cancérologie de Marseille, CRCM, TrGET Pre-Clinical Assay Platform, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Yuji Yamanashi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Daniel Olive
- Centre de Recherche en Cancérologie de Marseille, CRCM, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Rémy Castellano
- Centre de Recherche en Cancérologie de Marseille, CRCM, TrGET Pre-Clinical Assay Platform, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Geoffrey Guittard
- Centre de Recherche en Cancérologie de Marseille, CRCM, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Jacques A Nunès
- Centre de Recherche en Cancérologie de Marseille, CRCM, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France.
| |
Collapse
|
5
|
Purcarea A, Jarosch S, Barton J, Grassmann S, Pachmayr L, D'Ippolito E, Hammel M, Hochholzer A, Wagner KI, van den Berg JH, Buchholz VR, Haanen JBAG, Busch DH, Schober K. Signatures of recent activation identify a circulating T cell compartment containing tumor-specific antigen receptors with high avidity. Sci Immunol 2022; 7:eabm2077. [PMID: 35960818 DOI: 10.1126/sciimmunol.abm2077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
T cell receptor (TCR) avidity is assumed to be a major determinant of the spatiotemporal fate and protective capacity of tumor-specific T cells. However, monitoring polyclonal T cell responses with known TCR avidities in vivo over space and time remains challenging. Here, we investigated the fate and functionality of tumor neoantigen-specific T cells with TCRs of distinct avidities in a well-established, reductionist preclinical tumor model and human patients with melanoma. To this end, we used polyclonal T cell transfers with in-depth characterized TCRs together with flow cytometric phenotyping in mice inoculated with MC38 OVA tumors. Transfer of T cells from retrogenic mice harboring TCRs with high avidity resulted in best tumor protection. Unexpectedly, we found that both high- and low-avidity T cells are similarly abundant within the tumor and adopt concordant phenotypic signs of exhaustion. Outside the tumor, high-avidity TCR T cells were not generally overrepresented but, instead, selectively enriched in T cell populations with intermediate PD-1 protein expression. Single-cell sequencing of neoantigen-specific T cells from two patients with melanoma-combined with transgenic reexpression of identified TCRs by CRISPR-Cas9-mediated orthotopic TCR replacement-revealed high-functionality TCRs to be enriched in T cells with RNA signatures of recent activation. Furthermore, of 130 surface protein candidates, PD-1 surface expression was most consistently enriched in functional TCRs. Together, our findings show that tumor-reactive TCRs with high protective capacity circulating in peripheral blood are characterized by a signature of recent activation.
Collapse
Affiliation(s)
- Anna Purcarea
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Jack Barton
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Simon Grassmann
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Ludwig Pachmayr
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Elvira D'Ippolito
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Monika Hammel
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Anna Hochholzer
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Karolin I Wagner
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | | | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - John B A G Haanen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany.,Focus Group "Clinical Cell Processing and Purification", Institute for Advanced Study, TUM, Munich, Germany
| | - Kilian Schober
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany.,Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie, und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
6
|
Füchsl F, Krackhardt AM. Adoptive Cellular Therapy for Multiple Myeloma Using CAR- and TCR-Transgenic T Cells: Response and Resistance. Cells 2022; 11:410. [PMID: 35159220 PMCID: PMC8834324 DOI: 10.3390/cells11030410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/15/2022] Open
Abstract
Despite the substantial improvement of therapeutic approaches, multiple myeloma (MM) remains mostly incurable. However, immunotherapeutic and especially T cell-based approaches pioneered the therapeutic landscape for relapsed and refractory disease recently. Targeting B-cell maturation antigen (BCMA) on myeloma cells has been demonstrated to be highly effective not only by antibody-derived constructs but also by adoptive cellular therapies. Chimeric antigen receptor (CAR)-transgenic T cells lead to deep, albeit mostly not durable responses with manageable side-effects in intensively pretreated patients. The spectrum of adoptive T cell-transfer covers synthetic CARs with diverse specificities as well as currently less well-established T cell receptor (TCR)-based personalized strategies. In this review, we want to focus on treatment characteristics including efficacy and safety of CAR- and TCR-transgenic T cells in MM as well as the future potential these novel therapies may have. ACT with transgenic T cells has only entered clinical trials and various engineering strategies for optimization of T cell responses are necessary to overcome therapy resistance mechanisms. We want to outline the current success in engineering CAR- and TCR-T cells, but also discuss challenges including resistance mechanisms of MM for evading T cell therapy and point out possible novel strategies.
Collapse
Affiliation(s)
- Franziska Füchsl
- School of Medicine, Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany;
| | - Angela M. Krackhardt
- School of Medicine, Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, Technische Universität München, Ismaningerstraße 22, 81675 Munich, Germany;
- German Cancer Consortium (DKTK), Partner-Site Munich, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Einsteinstraße 25, 81675 Munich, Germany
| |
Collapse
|
7
|
Edmunds GL, Wong CCW, Ambler R, Milodowski EJ, Alamir H, Cross SJ, Galea G, Wülfing C, Morgan DJ. Adenosine 2A receptor and TIM3 suppress cytolytic killing of tumor cells via cytoskeletal polarization. Commun Biol 2022; 5:9. [PMID: 35013519 PMCID: PMC8748690 DOI: 10.1038/s42003-021-02972-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022] Open
Abstract
Tumors generate an immune-suppressive environment that prevents effective killing of tumor cells by CD8+ cytotoxic T cells (CTL). It remains largely unclear upon which cell type and at which stage of the anti-tumor response mediators of suppression act. We have combined an in vivo tumor model with a matching in vitro reconstruction of the tumor microenvironment based on tumor spheroids to identify suppressors of anti-tumor immunity that directly act on interaction between CTL and tumor cells and to determine mechanisms of action. An adenosine 2A receptor antagonist, as enhanced by blockade of TIM3, slowed tumor growth in vivo. Engagement of the adenosine 2A receptor and TIM3 reduced tumor cell killing in spheroids, impaired CTL cytoskeletal polarization ex vivo and in vitro and inhibited CTL infiltration into tumors and spheroids. With this role in CTL killing, blocking A2AR and TIM3 may complement therapies that enhance T cell priming, e.g. anti-PD-1 and anti-CTLA-4.
Collapse
Affiliation(s)
- Grace L Edmunds
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Carissa C W Wong
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Rachel Ambler
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Hanin Alamir
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Stephen J Cross
- Wolfson BioImaging Facility, University of Bristol, Bristol, BS8 1TD, UK
| | - Gabriella Galea
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Christoph Wülfing
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
| | - David J Morgan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
8
|
Maeng HM, Moore BN, Bagheri H, Steinberg SM, Inglefield J, Dunham K, Wei WZ, Morris JC, Terabe M, England LC, Roberson B, Rosing D, Sachdev V, Pack SD, Miettinen MM, Barr FG, Weiner LM, Panch S, Stroncek DF, Wood LV, Berzofsky JA. Phase I Clinical Trial of an Autologous Dendritic Cell Vaccine Against HER2 Shows Safety and Preliminary Clinical Efficacy. Front Oncol 2021; 11:789078. [PMID: 34976830 PMCID: PMC8716407 DOI: 10.3389/fonc.2021.789078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/08/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Despite recent advances, there is an urgent need for agents targeting HER2-expressing cancers other than breast cancer. We report a phase I study (NCT01730118) of a dendritic cell (DC) vaccine targeting HER2 in patients with metastatic cancer or bladder cancer at high risk of relapse. PATIENTS AND METHODS Part 1 of the study enrolled patients with HER2-expressing metastatic cancer that had progressed after at least standard treatment and patients who underwent definitive treatment for invasive bladder cancer with no evidence of disease at the time of enrollment. Part 2 enrolled patients with HER2-expressing metastatic cancer who had progressed after anti-HER2 therapy. The DC vaccines were prepared from autologous monocytes and transduced with an adenoviral vector expressing the extracellular and transmembrane domains of HER2 (AdHER2). A total of five doses were planned, and adverse events were recorded in patients who received at least one dose. Objective response was evaluated by unidimensional immune-related response criteria every 8 weeks in patients who received at least two doses. Humoral and cellular immunogenicity were assessed in patients who received more than three doses. RESULTS A total of 33 patients were enrolled at four dose levels (5 × 106, 10 × 106, 20 × 106, and 40 × 106 DCs). Median follow-up duration was 36 weeks (4-124); 10 patients completed five doses. The main reason for going off-study was disease progression. The main adverse events attributable to the vaccine were injection-site reactions. No cardiac toxicity was noted. Seven of 21 evaluable patients (33.3%) demonstrated clinical benefit (1 complete response, 1 partial response, and 5 stable disease). After ≥3 doses, an antibody response was detected in 3 of 13 patients (23.1%), including patients with complete and partial responses. Lymphocytes from 10 of 11 patients (90.9%) showed induction of anti-HER2 responses measured by the production of at least one of interferon-gamma, granzyme B, or tumor necrosis factor-alpha, and there were multifunctional responses in 8 of 11 patients (72.7%). CONCLUSIONS The AdHER2 DC vaccine showed evidence of immunogenicity and preliminary clinical benefit in patients with HER2-expressing cancers, along with an excellent safety profile. It shows promise for further clinical applications, especially in combination regimens.
Collapse
Affiliation(s)
- Hoyoung M. Maeng
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States,*Correspondence: Hoyoung M. Maeng,
| | - Brittni N. Moore
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Hadi Bagheri
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, Rockville, MD, United States
| | - Jon Inglefield
- Clinical Support Laboratory, Applied/Developmental Research Directorate, Frederick National Laboratory, Frederick, MD, United States
| | - Kim Dunham
- Clinical Support Laboratory, Applied/Developmental Research Directorate, Frederick National Laboratory, Frederick, MD, United States
| | - Wei-Zen Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - John C. Morris
- Division of Hematology-Oncology, University of Cincinnati, Cincinnati, OH, United States
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Lee C. England
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Brenda Roberson
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Douglas Rosing
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| | - Vandana Sachdev
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| | - Svetlana D. Pack
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Markku M. Miettinen
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Frederic G. Barr
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Louis M. Weiner
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - Sandhya Panch
- Center for Cellular Engineering, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - David F. Stroncek
- Center for Cellular Engineering, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Lauren V. Wood
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
9
|
Olguín-Contreras LF, Mendler AN, Popowicz G, Hu B, Noessner E. Double Strike Approach for Tumor Attack: Engineering T Cells Using a CD40L:CD28 Chimeric Co-Stimulatory Switch Protein for Enhanced Tumor Targeting in Adoptive Cell Therapy. Front Immunol 2021; 12:750478. [PMID: 34912334 PMCID: PMC8666660 DOI: 10.3389/fimmu.2021.750478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
Activation of co-stimulatory pathways in cytotoxic T lymphocytes expressing chimeric antigen receptors (CARs) have proven to boost effector activity, tumor rejection and long-term T cell persistence. When using antigen-specific T cell receptors (TCR) instead of CARs, the lack of co-stimulatory signals hampers robust antitumoral response, hence limiting clinical efficacy. In solid tumors, tumor stroma poses an additional hurdle through hindrance of infiltration and active inhibition. Our project aimed at generating chimeric co-stimulatory switch proteins (CSP) consisting of intracellular co-stimulatory domains (ICD) fused to extracellular protein domains (ECD) for which ligands are expressed in solid tumors. The ECD of CD40L was selected for combination with the ICD from the CD28 protein. With this approach, it was expected to not only provide co-stimulation and strengthen the TCR signaling, but also, through the CD40L ECD, facilitate the activation of tumor-resident antigen-presenting cells (APCs), modulate activation of tumor endothelium and induce TCR-MHC independent apoptotic effect on tumor cells. Since CD28 and CD40L belong to different classes of transmembrane proteins (type I and type II, respectively), creating a chimeric protein presented a structural and functional challenge. We present solutions to this challenge describing different CSP formats that were successfully expressed in human T cells along with an antigen-specific TCR. The level of surface expression of the CSPs depended on their distinct design and the state of T cell activation. In particular, CSPs were upregulated by TCR stimulation and downregulated following interaction with CD40 on target cells. Ligation of the CSP in the context of TCR-stimulation modulated intracellular signaling cascades and led to improved TCR-induced cytokine secretion and cytotoxicity. Moreover, the CD40L ECD exhibited activity as evidenced by effective maturation and activation of B cells and DCs. CD40L:CD28 CSPs are a new type of switch proteins designed to exert dual beneficial antitumor effect by acting directly on the gene-modified T cells and simultaneously on tumor cells and tumor-supporting cells of the TME. The observed effects suggest that they constitute a promising tool to be included in the engineering process of T cells to endow them with complementary features for improved performance in the tumor milieu.
Collapse
Affiliation(s)
| | - Anna N. Mendler
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany
| | - Grzegorz Popowicz
- Institute of Structural Biology, Helmholtz Center Munich, Munich, Germany
| | - Bin Hu
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany
- Immunoanalytics Research Group - Tissue Control of Immunocytes, Helmholtz Center Munich, Munich, Germany
| |
Collapse
|
10
|
Sugiyarto G, Prossor D, Dadas O, Arcia-Anaya ED, Elliott T, James E. Protective low-avidity anti-tumour CD8+ T cells are selectively attenuated by regulatory T cells. IMMUNOTHERAPY ADVANCES 2021; 1:ltaa001. [PMID: 33748824 PMCID: PMC7958313 DOI: 10.1093/immadv/ltaa001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives Regulatory T cells (Treg) play a major role in the suppression of protective anti-tumour T cell responses. In the CT26 BALB/c murine model of colorectal carcinoma, Tregs differentially suppress responses to two characterised CD8+ T epitopes, AH1 and GSW11, which results in an absence of detectable IFN-γ-producing GSW11-specific T cells in the spleen and lymph nodes of tumour challenged mice. Activation of GSW11-specific T cells correlates with protection against tumour progression. We wanted to examine the presence of non-functional GSW11-specific T cells in Treg replete and depleted mice, assess their phenotype and their affinity compared to AH1-specific T cells. Methods We used peptide-specific tetramers to identify tumour-specific CD8+ T cells and assessed the cell surface expression of markers associated with exhaustion (PD-1, Tim3 and Lag-3) and their function by IFN-g production using flow cytometry. We also assessed the T cell receptor (TcR) clonality of tumour-specific T cells. Tetramer competition assays were performed to determine the relative affinity of identified TcR. Results Here, we show that GSW11-specific T cells are in fact induced in Treg-replete, CT26-bearing mice, where they make up the majority of tumour-infiltrating CD8+ lymphocytes, but exhibit an ‘exhausted’ phenotype. This dysfunctional phenotype is induced early in the anti-tumour response in tumours. Depletion of Tregs prior to tumour challenge correlates with an altered T cell receptor (TcR) repertoire. Moreover, the avidity of GSW11-specific TcRs that expanded in the absence of Tregs was significantly lower compared with TcRs of CD8+populations that were diminished in protective anti-tumour responses. Conclusion Our results indicate that Tregs suppress the induction of protective anti-tumour T cell responses and may signify that low-avidity T cells play an important role in this protection.
Collapse
Affiliation(s)
- Gessa Sugiyarto
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - David Prossor
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Osman Dadas
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - E David Arcia-Anaya
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Tim Elliott
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,Institute for Life sciences, University of Southampton, Southampton, UK
| | - Edward James
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,Institute for Life sciences, University of Southampton, Southampton, UK
| |
Collapse
|
11
|
Kim SI, Cassella CR, Byrne KT. Tumor Burden and Immunotherapy: Impact on Immune Infiltration and Therapeutic Outcomes. Front Immunol 2021; 11:629722. [PMID: 33597954 PMCID: PMC7882695 DOI: 10.3389/fimmu.2020.629722] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy has revolutionized the treatment landscape in medical oncology, but its efficacy has been variable across patients. Biomarkers to predict such differential response to immunotherapy include cytotoxic T lymphocyte infiltration, tumor mutational burden, and microsatellite instability. A growing number of studies also suggest that baseline tumor burden, or tumor size, predicts response to immunotherapy. In this review, we discuss the changes in immune profile and therapeutic responses that occur with increasing tumor size. We also overview therapeutic approaches to reduce tumor burden and favorably modulate the immune microenvironment of larger tumors.
Collapse
Affiliation(s)
- Samuel I Kim
- Program in Biochemistry, College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher R Cassella
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katelyn T Byrne
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Castro-Sanchez P, Teagle AR, Prade S, Zamoyska R. Modulation of TCR Signaling by Tyrosine Phosphatases: From Autoimmunity to Immunotherapy. Front Cell Dev Biol 2020; 8:608747. [PMID: 33425916 PMCID: PMC7793860 DOI: 10.3389/fcell.2020.608747] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Early TCR signaling is dependent on rapid phosphorylation and dephosphorylation of multiple signaling and adaptor proteins, leading to T cell activation. This process is tightly regulated by an intricate web of interactions between kinases and phosphatases. A number of tyrosine phosphatases have been shown to modulate T cell responses and thus alter T cell fate by negatively regulating early TCR signaling. Mutations in some of these enzymes are associated with enhanced predisposition to autoimmunity in humans, and mouse models deficient in orthologous genes often show T cell hyper-activation. Therefore, phosphatases are emerging as potential targets in situations where it is desirable to enhance T cell responses, such as immune responses to tumors. In this review, we summarize the current knowledge about tyrosine phosphatases that regulate early TCR signaling and discuss their involvement in autoimmunity and their potential as targets for tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Rose Zamoyska
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Fischbeck AJ, Ruehland S, Ettinger A, Paetzold K, Masouris I, Noessner E, Mendler AN. Tumor Lactic Acidosis: Protecting Tumor by Inhibiting Cytotoxic Activity Through Motility Arrest and Bioenergetic Silencing. Front Oncol 2020; 10:589434. [PMID: 33364193 PMCID: PMC7753121 DOI: 10.3389/fonc.2020.589434] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Adoptive T cell therapy (ACT) is highly effective in the treatment of hematologic malignancies, but shows limited success in solid tumors. Inactivation of T cells in the tumor milieu is a major hurdle to a wider application of ACT. Cytotoxicity is the most relevant activity for tumor eradication. Here, we document that cytotoxic T cells (CTL) in lactic acidosis exhibited strongly reduced tumor cell killing, which could be compensated partly by increasing the CTL to tumor cell ratio. Lactic acid intervened at multiple steps of the killing process. Lactic acid repressed the number of CTL that performed lytic granule exocytosis (degranulation) in tumor cell co-culture, and, additionally impaired the quality of the response, as judged by the reduced intensity of degranulation and lower secretion of cytotoxins (perforin, granzyme B, granzyme A). CTL in lactic acid switched to a low bioenergetic profile with an inability to metabolize glucose efficiently. They responded to anti-CD3 stimulation poorly with less extracellular acidification rate (ECAR). This might explain their repressed granule exocytosis activity. Using live cell imaging, we show that CTL in lactic acid have reduced motility, resulting in lower field coverage. Many CTL in lactic acidosis did not make contact with tumor cells; however, those which made contact, adhered to the tumor cell much longer than a CTL in normal medium. Reduced motility together with prolonged contact duration hinders serial killing, a defining feature of killing potency, but also locally confines cytotoxic activity, which helps to reduce the risk of collateral organ damage. These activities define lactic acid as a major signaling molecule able to orchestrate the spatial distribution of CTL inside inflamed tissue, such as cancer, as well as moderating their functional response. Lactic acid intervention and strategies to improve T cell metabolic fitness hold promise to improve the clinical efficacy of T cell–based cancer immunotherapy.
Collapse
Affiliation(s)
| | - Svenja Ruehland
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Biology II, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Andreas Ettinger
- Institute of Epigenetics and Stem Cells, Helmholtz Center Munich, Munich, Germany
| | | | - Ilias Masouris
- Immunoanalytics, Helmholtz Center Munich, Munich, Germany
| | | | - Anna N Mendler
- Immunoanalytics, Helmholtz Center Munich, Munich, Germany
| |
Collapse
|
14
|
Ambler R, Edmunds GL, Tan SL, Cirillo S, Pernes JI, Ruan X, Huete-Carrasco J, Wong CCW, Lu J, Ward J, Toti G, Hedges AJ, Dovedi SJ, Murphy RF, Morgan DJ, Wülfing C. PD-1 suppresses the maintenance of cell couples between cytotoxic T cells and target tumor cells within the tumor. Sci Signal 2020; 13:13/649/eaau4518. [PMID: 32934075 DOI: 10.1126/scisignal.aau4518] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The killing of tumor cells by CD8+ T cells is suppressed by the tumor microenvironment, and increased expression of inhibitory receptors, including programmed cell death protein-1 (PD-1), is associated with tumor-mediated suppression of T cells. To find cellular defects triggered by tumor exposure and associated PD-1 signaling, we established an ex vivo imaging approach to investigate the response of antigen-specific, activated effector CD8+ tumor-infiltrating lymphocytes (TILs) after interaction with target tumor cells. Although TIL-tumor cell couples readily formed, couple stability deteriorated within minutes. This was associated with impaired F-actin clearing from the center of the cellular interface, reduced Ca2+ signaling, increased TIL locomotion, and impaired tumor cell killing. The interaction of CD8+ T lymphocytes with tumor cell spheroids in vitro induced a similar phenotype, supporting a critical role of direct T cell-tumor cell contact. Diminished engagement of PD-1 within the tumor, but not acute ex vivo blockade, partially restored cell couple maintenance and killing. PD-1 thus contributes to the suppression of TIL function by inducing a state of impaired subcellular organization.
Collapse
Affiliation(s)
- Rachel Ambler
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Grace L Edmunds
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Sin Lih Tan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Silvia Cirillo
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Jane I Pernes
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Xiongtao Ruan
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jorge Huete-Carrasco
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Carissa C W Wong
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Jiahe Lu
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Juma Ward
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Giulia Toti
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Alan J Hedges
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Simon J Dovedi
- R&D Oncology, AstraZeneca, Granta Park, Cambridge CB21 6GH, UK
| | - Robert F Murphy
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,Departments of Biological Sciences, Biomedical Engineering and Machine Learning, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,Freiburg Institute for Advanced Studies and Faculty of Biology, Albert Ludwig University of Freiburg, 79104 Freiburg, Germany
| | - David J Morgan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK.
| | - Christoph Wülfing
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
15
|
Wang Z, Li K, Chen W, Wang X, Huang Y, Wang W, Wu W, Cai Z, Huang W. Modulation of SRSF2 expression reverses the exhaustion of TILs via the epigenetic regulation of immune checkpoint molecules. Cell Mol Life Sci 2020; 77:3441-3452. [PMID: 31838573 PMCID: PMC7426320 DOI: 10.1007/s00018-019-03362-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 10/15/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023]
Abstract
The elevated expression of immune checkpoints by the tumor microenvironment is associated with poor prognosis in several cancers due to the exhaustion of tumor-infiltrating lymphocytes (TILs), and the effective suppression of the expression of these genes is key to reversing the exhaustion of TILs. Herein, we determined that serine/arginine-rich splicing factor 2 (SRSF2) is a target for blocking the tumor microenvironment-associated immunosuppressive effects. We found that the expression of SRSF2 was increased in exhausted T cells and that SRSF2 was involved in multiple immune checkpoint molecules mediating TILs' exhaustion. Furthermore, SRSF2 was revealed to regulate the transcription of these immune checkpoint genes by associating with an acyl-transferases P300/CBP complex and altering the H3K27Ac level near these genes, thereafter influencing the recruitment of signal transducer and activator of transcription 3 (STAT3) to these gene promoters. Collectively, our data indicated that SRSF2 functions as a modulator of the anti-tumor response of T cells and may be a therapeutic target for reversing the exhaustion of TILs.
Collapse
Affiliation(s)
- Ziqiang Wang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China
| | - Kun Li
- Department of Nuclear Medicine, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, 250014, China
| | - Wei Chen
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China
| | - Xiaoxia Wang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China
| | - Yikun Huang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China
| | - Weiming Wang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China
| | - Wanjun Wu
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China
| | - Zhiming Cai
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China.
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China.
| | - Weiren Huang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, 518039, China.
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, 518035, China.
| |
Collapse
|
16
|
Snook JP, Soedel AJ, Ekiz HA, O'Connell RM, Williams MA. Inhibition of SHP-1 Expands the Repertoire of Antitumor T Cells Available to Respond to Immune Checkpoint Blockade. Cancer Immunol Res 2020; 8:506-517. [PMID: 32075800 DOI: 10.1158/2326-6066.cir-19-0690] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/12/2019] [Accepted: 02/11/2020] [Indexed: 12/13/2022]
Abstract
The presence and activity of CD8+ T cells within the tumor microenvironment are essential for the control of tumor growth. Utilizing B16-F10 melanoma tumors that express altered peptide ligands of chicken ovalbumin, OVA257-264, we measured high- and low-affinity OVA-specific responses following adoptive transfer of OT-I CD8+ T cell into mice subsequently challenged with tumors. T-cell receptor (TCR) affinity positively correlated with the frequency of OT-I tumor-infiltrating lymphocytes (TIL). Differences in TCR affinity inversely corresponded to in vivo tumor growth rate. Blockade of the PD-1 and CTLA-4 checkpoints preferentially increased the frequency and antitumor function of TIL responding to high-affinity antigens, while failing to enhance the antitumor activity of low-affinity T cells. To determine whether lowering the TCR activation threshold could enhance the breadth and magnitude of the antitumor T-cell response, we inhibited Src homology region 2 domain-containing phosphatase 1 (SHP-1) in OT-I T cells prior to tumor antigen exposure. SHP-1 knockdown increased the cytokine-producing potential of high- and low-affinity T cells but failed to enhance control of tumor growth. In contrast, when SHP-1 knockdown of OT-I T cells was combined with immunotherapy, we observed a significant and long-lasting suppression of tumor growth mediated by low-affinity T cells. We conclude that lowering the TCR activation threshold by targeting SHP-1 expands the repertoire of T cells available to respond to conventional checkpoint blockade, leading to enhanced control of tumor growth.
Collapse
Affiliation(s)
- Jeremy P Snook
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah
| | - Ashleigh J Soedel
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah
| | - H Atakan Ekiz
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah
| | - Matthew A Williams
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah. .,Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah
| |
Collapse
|
17
|
Ioannidou K, Randin O, Semilietof A, Maby-El Hajjami H, Baumgaertner P, Vanhecke D, Speiser DE. Low Avidity T Cells Do Not Hinder High Avidity T Cell Responses Against Melanoma. Front Immunol 2019; 10:2115. [PMID: 31555299 PMCID: PMC6742971 DOI: 10.3389/fimmu.2019.02115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/02/2022] Open
Abstract
The efficacy of T cells depends on their functional avidity, i. e., the strength of T cell interaction with cells presenting cognate antigen. The overall T cell response is composed of multiple T cell clonotypes, involving different T cell receptors and variable levels of functional avidity. Recently, it has been proposed that the presence of low avidity tumor antigen-specific CD8 T cells hinder their high avidity counterparts to protect from tumor growth. Here we analyzed human cytotoxic CD8 T cells specific for the melanoma antigen Melan-A/MART-1. We found that the presence of low avidity T cells did not result in reduced cytotoxicity of tumor cells, nor reduced cytokine production, by high avidity T cells. In vivo in NSG-HLA-A2 mice, the anti-tumor effect of high avidity T cells was similar in presence or absence of low avidity T cells. These data indicate that low avidity T cells are not hindering anti-tumor T cell responses, a finding that is reassuring because low avidity T cells are an integrated part of natural T cell responses.
Collapse
Affiliation(s)
- Kalliopi Ioannidou
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Olivier Randin
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Aikaterini Semilietof
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Hélène Maby-El Hajjami
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Petra Baumgaertner
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Dominique Vanhecke
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| |
Collapse
|
18
|
Cirillo N, Morgan DJ, Pedicillo MC, Celentano A, Lo Muzio L, McCullough MJ, Prime SS. Characterisation of the cancer-associated glucocorticoid system: key role of 11β-hydroxysteroid dehydrogenase type 2. Br J Cancer 2017; 117:984-993. [PMID: 28797028 PMCID: PMC5625663 DOI: 10.1038/bjc.2017.243] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 04/11/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Recent studies have shown that production of cortisol not only takes place in several non-adrenal peripheral tissues such as epithelial cells but, also, the local inter-conversion between cortisone and cortisol is regulated by the 11β-hydroxysteroid dehydrogenases (11β-HSDs). However, little is known about the activity of this non-adrenal glucocorticoid system in cancers. Methods: The presence of a functioning glucocorticoid system was assessed in human skin squamous cell carcinoma (SCC) and melanoma and further, in 16 epithelial cell lines from 8 different tissue types using ELISA, western blotting and immunofluorescence. 11β-HSD2 was inhibited both pharmacologically and by siRNA technology. Naïve CD8+ T cells were used to test the paracrine effects of cancer-derived cortisol on the immune system in vitro. Functional assays included cell–cell adhesion and cohesion in two- and three-dimensional models. Immunohistochemical data of 11β-HSD expression were generated using tissue microarrays of 40 cases of human SCCs as well as a database featuring 315 cancer cases from 15 different tissues. Results: We show that cortisol production is a common feature of malignant cells and has paracrine functions. Cortisol production correlated with the magnitude of glucocorticoid receptor (GR)-dependent inhibition of tumour-specific CD8+ T cells in vitro. 11β-HSDs were detectable in human skin SCCs and melanoma. Analyses of publicly available protein expression data of 11β-HSDs demonstrated that 11β-HSD1 and -HSD2 were dysregulated in the majority (73%) of malignancies. Pharmacological manipulation of 11β-HSD2 activity by 18β-glycyrrhetinic acid (GA) and silencing by specific siRNAs modulated the bioavailability of cortisol. Cortisol also acted in an autocrine manner and promoted cell invasion in vitro and cell–cell adhesion and cohesion in two- and three-dimensional models. Immunohistochemical analyses using tissue microarrays showed that expression of 11β-HSD2 was significantly reduced in human SCCs of the skin. Conclusions: The results demonstrate evidence of a cancer-associated glucocorticoid system and show for the first time, the functional significance of cancer-derived cortisol in tumour progression.
Collapse
Affiliation(s)
- Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, Melbourne, VIC 3053, Australia
| | - David J Morgan
- School of Cellular &Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | | | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, Melbourne, VIC 3053, Australia
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71122, Italy
| | - Michael J McCullough
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, Melbourne, VIC 3053, Australia
| | - Stephen S Prime
- Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK
| |
Collapse
|
19
|
Bruns M, Deppert W. Immunotherapy of WAP-T NP mice with early stage mammary gland tumors. Oncotarget 2017; 8:67790-67804. [PMID: 28978072 PMCID: PMC5620212 DOI: 10.18632/oncotarget.18850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/05/2017] [Indexed: 12/27/2022] Open
Abstract
The SV40 transgenic BALB/c mouse based WAP-T/WAP-TNP model for triple-negative breast cancer allows the analysis of parameters influencing immunotherapeutic approaches. Except for WAP-TNP tumors expressing the immune-dominant LCMV NP-epitope within SV40 T-antigen (T-AgNP) which is not expressed by T-Ag of WAP-T tumors, the tumors are extremely similar. Comparative anti-PD1/PD-L1 immunotherapy of WAP-T and WAP-TNP mice supported the hypothesis that the immunogenicity of tumor antigen T-cell epitopes strongly influences the success of immune checkpoint blockade therapy, with highly immunogenic T-cell epitopes favoring rapid CTL exhaustion. Here we analyzed the immune response in NP8 mice during early times of tumor development. LCMV infection of lactating NP8 mice induced lifelong tumor protection by memory CTLs. Immunization with LCMV after involution and appearance of T-AgNP expressing parity-induced tumor progenitor cells could not cure the mice, as memory CTLs became exhausted. However, immunization significantly prolonged the time of tumor outgrowth. Elimination of exhausted CTLs and of immunosuppressive cells by sub-lethal γ-irradiation, followed by adoptive transfer of NP-epitope specific CTLs into NP8 tumor mice with early lesions, completely prevented tumor outgrowth, when lymphocytes obtained after injection of weakly immunogenic NP8 tumor-derived cells into BALB/c mice were transferred. Transfer of lymphocytes obtained after infection of BALB/c mice with highly immunogenic LCMV into such mice delayed tumor outgrowth for a significant period, but could not prevent it. We conclude that eliminating exhausted CTLs and immune-suppressive cells followed by transfer or generation of low-avidity tumor antigen-specific CTLs might be a promising approach for curative tumor immunotherapy.
Collapse
Affiliation(s)
- Michael Bruns
- Heinrich-Pette-Institute, Leibniz-Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Wolfgang Deppert
- Heinrich-Pette-Institute, Leibniz-Institute for Experimental Virology, 20251 Hamburg, Germany.,Institute for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), University of Hamburg, 20246 Hamburg, Germany
| |
Collapse
|
20
|
Zahm CD, Colluru VT, McNeel DG. Vaccination with High-Affinity Epitopes Impairs Antitumor Efficacy by Increasing PD-1 Expression on CD8 + T Cells. Cancer Immunol Res 2017. [PMID: 28634215 DOI: 10.1158/2326-6066.cir-16-0374] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antitumor vaccines encoding self-antigens generally have low immunogenicity in clinical trials. Several approaches are aimed at improving vaccine immunogenicity, including efforts to alter encoded epitopes. Immunization with epitopes altered for increased affinity for the major histocompatibility complex (MHC) or T-cell receptor (TCR) elicits greater numbers of CD8 T cells but inferior antitumor responses. Our previous results suggested that programmed death 1 (PD-1) and its ligand (PD-L1) increased on antigen-specific CD8 T cells and tumor cells, respectively, after high-affinity vaccination. In this report, we use two murine models to investigate whether the dose, MHC affinity, or TCR affinity of an epitope affected the antitumor response via the PD-1/PD-L1 axis. T cells activated with high-affinity epitopes resulted in prolonged APC:T-cell contact time that led to elevated, persistent PD-1 expression, and expression of other checkpoint molecules, in vitro and in vivo Immunization with high-affinity epitopes also decreased antitumor efficacy in the absence of PD-1 blockade. Thus, APC:T-cell contact time can be altered by epitope affinity and lead to therapeutically relevant changes in vaccine efficacy mediated by changes in PD-1 expression. These findings have implications for the use of agents targeting PD-1 expression or function whenever high-affinity CD8 T cells are elicited or supplied by means of vaccination or adoptive transfer. Cancer Immunol Res; 5(8); 630-41. ©2017 AACR.
Collapse
Affiliation(s)
- Christopher D Zahm
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Viswa T Colluru
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin.
| |
Collapse
|
21
|
Schlenker R, Olguín-Contreras LF, Leisegang M, Schnappinger J, Disovic A, Rühland S, Nelson PJ, Leonhardt H, Harz H, Wilde S, Schendel DJ, Uckert W, Willimsky G, Noessner E. Chimeric PD-1:28 Receptor Upgrades Low-Avidity T cells and Restores Effector Function of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy. Cancer Res 2017; 77:3577-3590. [PMID: 28533272 DOI: 10.1158/0008-5472.can-16-1922] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/13/2016] [Accepted: 05/08/2017] [Indexed: 11/16/2022]
Abstract
Inherent intermediate- to low-affinity T-cell receptors (TCR) that develop during the natural course of immune responses may not allow sufficient activation for tumor elimination, making the majority of T cells suboptimal for adoptive T-cell therapy (ATT). TCR affinity enhancement has been implemented to provide stronger T-cell activity but carries the risk of creating undesired cross-reactivity leading to potential serious adverse effects in clinical application. We demonstrate here that engineering of low-avidity T cells recognizing a naturally processed and presented tumor-associated antigen with a chimeric PD-1:28 receptor increases effector function to levels seen with high-avidity T cells of identical specificity. Upgrading the function of low-avidity T cells without changing the TCR affinity will allow a large arsenal of low-avidity T cells previously thought to be therapeutically inefficient to be considered for ATT. PD-1:28 engineering reinstated Th1 function in tumor-infiltrating lymphocytes that had been functionally disabled in the human renal cell carcinoma environment without unleashing undesired Th2 cytokines or IL10. Involved mechanisms may be correlated to restoration of ERK and AKT signaling pathways. In mouse tumor models of ATT, PD-1:28 engineering enabled low-avidity T cells to proliferate stronger and prevented PD-L1 upregulation and Th2 polarization in the tumor milieu. Engineered T cells combined with checkpoint blockade secreted significantly more IFNγ compared with T cells without PD-1:28, suggesting a beneficial combination with checkpoint blockade therapy or other therapeutic strategies. Altogether, the supportive effects of PD-1:28 engineering on T-cell function make it an attractive tool for ATT. Cancer Res; 77(13); 3577-90. ©2017 AACR.
Collapse
Affiliation(s)
- Ramona Schlenker
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany
| | - Luis Felipe Olguín-Contreras
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany.,Immunoanalytics Research Group Tissue Control of Immunocytes & Core Facility, Helmholtz Center Munich, Munich, Germany
| | - Matthias Leisegang
- Institute of Immunology, Charité, Campus Buch, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Julia Schnappinger
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany.,Immunoanalytics Research Group Tissue Control of Immunocytes & Core Facility, Helmholtz Center Munich, Munich, Germany
| | - Anja Disovic
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany.,Immunoanalytics Research Group Tissue Control of Immunocytes & Core Facility, Helmholtz Center Munich, Munich, Germany
| | - Svenja Rühland
- Ludwig-Maximilian University Munich, Medizinische Klinik und Poliklinik IV, Munich, Germany.,Department of Biology II and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilian University Munich, Munich, Germany
| | - Peter J Nelson
- Ludwig-Maximilian University Munich, Medizinische Klinik und Poliklinik IV, Munich, Germany
| | - Heinrich Leonhardt
- Department of Biology II and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilian University Munich, Munich, Germany
| | - Hartmann Harz
- Department of Biology II and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilian University Munich, Munich, Germany
| | | | | | - Wolfgang Uckert
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Institute of Biology, Humboldt University Berlin, Berlin, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité, Campus Buch, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany. .,Immunoanalytics Research Group Tissue Control of Immunocytes & Core Facility, Helmholtz Center Munich, Munich, Germany
| |
Collapse
|
22
|
Noessner E. DGK-α: A Checkpoint in Cancer-Mediated Immuno-Inhibition and Target for Immunotherapy. Front Cell Dev Biol 2017; 5:16. [PMID: 28316970 PMCID: PMC5335622 DOI: 10.3389/fcell.2017.00016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/17/2017] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy is moving to the forefront of cancer treatments owing to impressive durable responses achieved with checkpoint blockade antibodies and adoptive T-cell therapy. Still, improvements are necessary since, overall, only a small percentage of patients benefit from current therapies. Here, I summarize evidence that DGK-α may represent an immunological checkpoint suppressing the activity of cytotoxic immunocytes in the tumor microenvironment. DGK-inhibitors can restore the antitumor function of tumor-suppressed adaptive and innate cytotoxic immunocytes. The activity of DGK-inhibitors lays downstream of current checkpoint blockade antibodies. Thus, synergistic effects are expected from combination strategies. Moreover, DGK-inhibitors may permit a double-strike attack on tumor cells as DGK-inhibition may not only re-instate immunological tumor attack but also may harm tumor cells directly by interfering with oncogenic survival pathways. Together, DGK-inhibitors have very promising characteristics and may be beneficially included into the armamentarium of cancer immunotherapeutics.
Collapse
Affiliation(s)
- Elfriede Noessner
- Immunoanalytics Core Facility and Research Group Tissue Control of Immunocytes, Helmholtz Zentrum München München, Germany
| |
Collapse
|
23
|
Diniz MO, Sales NS, Silva JR, Ferreira LCS. Protection against HPV-16-Associated Tumors Requires the Activation of CD8+ Effector Memory T Cells and the Control of Myeloid-Derived Suppressor Cells. Mol Cancer Ther 2016; 15:1920-30. [PMID: 27222537 DOI: 10.1158/1535-7163.mct-15-0742] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/10/2016] [Indexed: 11/16/2022]
Abstract
Active anticancer immunotherapeutic approaches have been shown to induce cellular or humoral immune responses in patients, but, thus far, the observed outcomes did not ensure their recommendation for clinical use. The induction of tumor-specific CD8(+) T cells, although required for the clearance of most solid tumors, was shown to be insufficient for the development of a successful immunotherapeutic approach. The suppressive immune environment triggered by tumors, including the expansion of myeloid-derived suppressor cells (MDSC), is detrimental to the development of antitumor immune responses and precludes the generation of more promising clinical outcomes. In this work, we characterized the CD8(+) T-cell population specifically involved in the control of tumor growth and the role of MDSCs after administration of an antitumor therapeutic DNA vaccine targeting human papillomavirus type 16 (HPV-16)-associated tumors. Activation of cytotoxic high-avidity CD8(+) T cells with an effector memory phenotype was found in mice grafted with tumor cells expressing the HPV-16 oncoproteins. In addition, MDSC antibody depletion further enhanced the immunotherapeutic effects of the vaccine, resulting in the complete eradication of tumor cells. Collectively, the current results indicate that the simultaneous control of MDSCs and activation of high-avidity tumor-specific effector memory CD8(+) T cells are key features for tumor protection by immunotherapeutic approaches and deserve further testing under clinical conditions. Mol Cancer Ther; 15(8); 1920-30. ©2016 AACR.
Collapse
Affiliation(s)
- Mariana O Diniz
- Department of Microbiology, Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Natiely S Sales
- Department of Microbiology, Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jamile R Silva
- Department of Microbiology, Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luís Carlos S Ferreira
- Department of Microbiology, Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
24
|
Allen CT, Clavijo PE, Van Waes C, Chen Z. Anti-Tumor Immunity in Head and Neck Cancer: Understanding the Evidence, How Tumors Escape and Immunotherapeutic Approaches. Cancers (Basel) 2015; 7:2397-414. [PMID: 26690220 PMCID: PMC4695900 DOI: 10.3390/cancers7040900] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/10/2015] [Accepted: 11/30/2015] [Indexed: 12/22/2022] Open
Abstract
Many carcinogen- and human papilloma virus (HPV)-associated head and neck cancers (HNSCC) display a hematopoietic cell infiltrate indicative of a T-cell inflamed phenotype and an underlying anti-tumor immune response. However, by definition, these tumors have escaped immune elimination and formed a clinically significant malignancy. A number of both genetic and environmental mechanisms may allow such immune escape, including selection of poorly antigenic cancer cell subsets, tumor produced proinflammatory and immunosuppressive cytokines, recruitment of immunosuppressive immune cell subsets into the tumor and expression of checkpoint pathway components that limit T-cell responses. Here, we explore concepts of antigenicity and immunogenicity in solid tumors, summarize the scientific and clinical data that supports the use of immunotherapeutic approaches in patients with head and neck cancer, and discuss immune-based treatment approaches currently in clinical trials.
Collapse
Affiliation(s)
- Clint T Allen
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
| | - Paul E Clavijo
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Carter Van Waes
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Zhong Chen
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Caruso HG, Hurton LV, Najjar A, Rushworth D, Ang S, Olivares S, Mi T, Switzer K, Singh H, Huls H, Lee DA, Heimberger AB, Champlin RE, Cooper LJN. Tuning Sensitivity of CAR to EGFR Density Limits Recognition of Normal Tissue While Maintaining Potent Antitumor Activity. Cancer Res 2015; 75:3505-18. [PMID: 26330164 DOI: 10.1158/0008-5472.can-15-0139] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Many tumors overexpress tumor-associated antigens relative to normal tissue, such as EGFR. This limits targeting by human T cells modified to express chimeric antigen receptors (CAR) due to potential for deleterious recognition of normal cells. We sought to generate CAR(+) T cells capable of distinguishing malignant from normal cells based on the disparate density of EGFR expression by generating two CARs from monoclonal antibodies that differ in affinity. T cells with low-affinity nimotuzumab-CAR selectively targeted cells overexpressing EGFR, but exhibited diminished effector function as the density of EGFR decreased. In contrast, the activation of T cells bearing high-affinity cetuximab-CAR was not affected by the density of EGFR. In summary, we describe the generation of CARs able to tune T-cell activity to the level of EGFR expression in which a CAR with reduced affinity enabled T cells to distinguish malignant from nonmalignant cells.
Collapse
Affiliation(s)
- Hillary G Caruso
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Lenka V Hurton
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Amer Najjar
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Rushworth
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Sonny Ang
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simon Olivares
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tiejuan Mi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kirsten Switzer
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Harjeet Singh
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Helen Huls
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dean A Lee
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laurence J N Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas.
| |
Collapse
|
26
|
Hebeisen M, Allard M, Gannon PO, Schmidt J, Speiser DE, Rufer N. Identifying Individual T Cell Receptors of Optimal Avidity for Tumor Antigens. Front Immunol 2015; 6:582. [PMID: 26635796 PMCID: PMC4649060 DOI: 10.3389/fimmu.2015.00582] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/30/2015] [Indexed: 02/02/2023] Open
Abstract
Cytotoxic T cells recognize, via their T cell receptors (TCRs), small antigenic peptides presented by the major histocompatibility complex (pMHC) on the surface of professional antigen-presenting cells and infected or malignant cells. The efficiency of T cell triggering critically depends on TCR binding to cognate pMHC, i.e., the TCR–pMHC structural avidity. The binding and kinetic attributes of this interaction are key parameters for protective T cell-mediated immunity, with stronger TCR–pMHC interactions conferring superior T cell activation and responsiveness than weaker ones. However, high-avidity TCRs are not always available, particularly among self/tumor antigen-specific T cells, most of which are eliminated by central and peripheral deletion mechanisms. Consequently, systematic assessment of T cell avidity can greatly help distinguishing protective from non-protective T cells. Here, we review novel strategies to assess TCR–pMHC interaction kinetics, enabling the identification of the functionally most-relevant T cells. We also discuss the significance of these technologies in determining which cells within a naturally occurring polyclonal tumor-specific T cell response would offer the best clinical benefit for use in adoptive therapies, with or without T cell engineering.
Collapse
Affiliation(s)
- Michael Hebeisen
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Mathilde Allard
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Philippe O Gannon
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Julien Schmidt
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland ; TCMetrix Sàrl , Epalinges , Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland ; Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland ; Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland
| |
Collapse
|
27
|
Palmer DC, Guittard GC, Franco Z, Crompton JG, Eil RL, Patel SJ, Ji Y, Van Panhuys N, Klebanoff CA, Sukumar M, Clever D, Chichura A, Roychoudhuri R, Varma R, Wang E, Gattinoni L, Marincola FM, Balagopalan L, Samelson LE, Restifo NP. Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance. J Exp Med 2015; 212:2095-113. [PMID: 26527801 PMCID: PMC4647263 DOI: 10.1084/jem.20150304] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 09/11/2015] [Indexed: 01/17/2023] Open
Abstract
Palmer et al. find that Cish, a member of the SOCS family, is induced by TCR stimulation in CD8+ T cells and inhibits their functional avidity against tumor. The authors uncover a novel mechanism of suppression for a SOCS member. Improving the functional avidity of effector T cells is critical in overcoming inhibitory factors within the tumor microenvironment and eliciting tumor regression. We have found that Cish, a member of the suppressor of cytokine signaling (SOCS) family, is induced by TCR stimulation in CD8+ T cells and inhibits their functional avidity against tumors. Genetic deletion of Cish in CD8+ T cells enhances their expansion, functional avidity, and cytokine polyfunctionality, resulting in pronounced and durable regression of established tumors. Although Cish is commonly thought to block STAT5 activation, we found that the primary molecular basis of Cish suppression is through inhibition of TCR signaling. Cish physically interacts with the TCR intermediate PLC-γ1, targeting it for proteasomal degradation after TCR stimulation. These findings establish a novel targetable interaction that regulates the functional avidity of tumor-specific CD8+ T cells and can be manipulated to improve adoptive cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yun Ji
- National Cancer Institute, Bethesda, MD 20892
| | | | | | | | - David Clever
- National Cancer Institute, Bethesda, MD 20892 Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, OH 43210
| | | | | | - Rajat Varma
- National Institute of Allergy and Infectious Disease, Bethesda, MD 20892
| | - Ena Wang
- Sidra Medical and Research Center, Doha, Qatar
| | | | | | | | | | | |
Collapse
|
28
|
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
|
29
|
Glass R, Synowitz M. CNS macrophages and peripheral myeloid cells in brain tumours. Acta Neuropathol 2014; 128:347-62. [PMID: 24722970 DOI: 10.1007/s00401-014-1274-2] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/10/2014] [Accepted: 03/25/2014] [Indexed: 12/15/2022]
Abstract
Primary brain tumours (gliomas) initiate a strong host response and can contain large amounts of immune cells (myeloid cells) such as microglia and tumour-infiltrating macrophages. In gliomas the course of pathology is not only controlled by the genetic make-up of the tumour cells, but also depends on the interplay with myeloid cells in the tumour microenvironment. Especially malignant gliomas such as glioblastoma multiforme (GBM) are notoriously immune-suppressive and it is now evident that GBM cells manipulate myeloid cells to support tumour expansion. The protumorigenic effects of glioma-associated myeloid cells comprise a support for angiogenesis as well as tumour cell invasion, proliferation and survival. Different strategies for inhibiting the pathological functions of myeloid cells in gliomas are explored, and blocking the tropism of microglia/macrophages to gliomas or manipulating the signal transduction pathways for immune cell activation has been successful in pre-clinical models. Hence, myeloid cells are now emerging as a promising target for new adjuvant therapies for gliomas. However, it is also becoming evident that some myeloid-directed glioma therapies may only be beneficial for distinct subclasses of gliomas and that a more cell-type-specific manipulation of either microglia or macrophages may improve therapeutic outcomes.
Collapse
|
30
|
Boldison J, Chu CJ, Copland DA, Lait PJP, Khera TK, Dick AD, Nicholson LB. Tissue-resident exhausted effector memory CD8+ T cells accumulate in the retina during chronic experimental autoimmune uveoretinitis. THE JOURNAL OF IMMUNOLOGY 2014; 192:4541-50. [PMID: 24740509 PMCID: PMC4009498 DOI: 10.4049/jimmunol.1301390] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Experimental autoimmune uveoretinitis is a model for noninfectious posterior segment intraocular inflammation in humans. Although this disease is CD4+ T cell dependent, in the persistent phase of disease CD8+ T cells accumulate. We show that these are effector memory CD8+ T cells that differ from their splenic counterparts with respect to surface expression of CD69, CD103, and Ly6C. These retinal effector memory CD8+ T cells have limited cytotoxic effector function, are impaired in their ability to proliferate in response to Ag-specific stimulation, and upregulate programmed death 1 receptor. Treatment with fingolimod (FTY720) during the late phase of disease revealed that retinal CD8+ T cells were tissue resident. Despite signs of exhaustion, these cells were functional, as their depletion resulted in an expansion of retinal CD4+ T cells and CD11b+ macrophages. These results demonstrate that, during chronic autoimmune inflammation, exhausted CD8+ T cells become established in the local tissue. They are phenotypically distinct from peripheral CD8+ T cells and provide local signals within the tissue by expression of inhibitory receptors such as programmed death 1 that limit persistent inflammation.
Collapse
Affiliation(s)
- Joanne Boldison
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
31
|
T cell avidity and tumor immunity: problems and solutions. CANCER MICROENVIRONMENT 2013; 7:1-9. [PMID: 24357332 DOI: 10.1007/s12307-013-0143-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/05/2013] [Indexed: 12/16/2022]
Abstract
A potent T cell response is an important component of durable anti-tumor immunity. The quality of the T cell response can, in-part, be measured by the avidity of the T cell for its tumor antigen-expressing target. While convention suggests that raising the avidity of the responding T cells may make for a more potent anti-tumor immune response, the threshold for effective tumor immunity remains unclear, as do some of the adverse effects of an inappropriately high avidity response. In this review, we discuss the relationship between T cell avidity and anti-tumor immunity, considering both experimental model systems as well as human clinical trials.
Collapse
|
32
|
Trick to treat: tricking the thymus to treat cancer. Blood 2013; 122:304-6. [PMID: 23869073 DOI: 10.1182/blood-2013-06-504100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
In this issue of Blood, Schmitt et al address the biology and safety of T cells engineered to express T-cell receptor (TCR) variants endowed with enhanced affinity for tumor-associated antigens.
Collapse
|
33
|
Dougan SK, Dougan M, Kim J, Turner JA, Ogata S, Cho HI, Jaenisch R, Celis E, Ploegh HL. Transnuclear TRP1-specific CD8 T cells with high or low affinity TCRs show equivalent antitumor activity. Cancer Immunol Res 2013; 1:99-111. [PMID: 24459675 PMCID: PMC3895912 DOI: 10.1158/2326-6066.cir-13-0047] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have generated, via somatic cell nuclear transfer, two independent lines of transnuclear (TN) mice, using as nuclear donors CD8 T cells, sorted by tetramer staining, that recognize the endogenous melanoma antigen TRP1. These two lines of nominally identical specificity differ greatly in their affinity for antigen (TRP1(high) or TRP1(low)) as inferred from tetramer dissociation and peptide responsiveness. Ex vivo-activated CD8 T cells from either TRP1(high) or TRP1(low) mice show cytolytic activity in 3D tissue culture and in vivo, and slow the progression of subcutaneous B16 melanoma. Although naïve TRP1(low) CD8 T cells do not affect tumor growth, upon activation these cells function indistinguishably from TRP1(high) cells in vivo, limiting tumor cell growth and increasing mouse survival. The anti-tumor effect of both TRP1(high) and TRP1(low) CD8 T cells is enhanced in RAG-deficient hosts. However, tumor outgrowth eventually occurs, likely due to T cell exhaustion. The TRP1 TN mice are an excellent model for examining the functional attributes of T cells conferred by TCR affinity, and they may serve as a platform for screening immunomodulatory cancer therapies.
Collapse
Affiliation(s)
- Stephanie K. Dougan
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142
| | - Michael Dougan
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Jun Kim
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142
- Massachusetts Institute of Technology, Cambridge, MA
| | - Jacob A. Turner
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142
- University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221
| | - Souichi Ogata
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142
- Janssen Research and Development, division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse B2340, Belgium
| | - Hyun-Il Cho
- Dept. of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142
| | - Esteban Celis
- Dept. of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612
| | - Hidde L. Ploegh
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142
| |
Collapse
|
34
|
Hailemichael Y, Dai Z, Jaffarzad N, Ye Y, Medina MA, Huang XF, Dorta-Estremera SM, Greeley NR, Nitti G, Peng W, Liu C, Lou Y, Wang Z, Ma W, Rabinovich B, Sowell RT, Schluns KS, Davis RE, Hwu P, Overwijk WW. Persistent antigen at vaccination sites induces tumor-specific CD8⁺ T cell sequestration, dysfunction and deletion. Nat Med 2013; 19:465-72. [PMID: 23455713 PMCID: PMC3618499 DOI: 10.1038/nm.3105] [Citation(s) in RCA: 356] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 01/25/2013] [Indexed: 12/22/2022]
Abstract
To understand why cancer vaccine-induced T cells often fail to eradicate tumors, we studied immune responses in mice vaccinated with gp100 melanoma peptide in incomplete Freund’s adjuvant (IFA), commonly used in clinical cancer vaccine trials. Peptide/IFA vaccination primed tumor-specific CD8+ T cells, which accumulated not in tumors but at the persisting, antigen-rich vaccination site. Once there, primed T cells became dysfunctional and underwent antigen-driven, Interferon-γ (IFN-γ) and Fas ligand (FasL)-mediated apoptosis, resulting in hyporesponsiveness to subsequent vaccination. Provision of anti-CD40 antibody, Toll-like receptor 7 (TLR7) agonist and interleukin-2 (IL-2) reduced T cell apoptosis but did not prevent vaccination site sequestration. A non-persisting vaccine formulation shifted T cell localization towards tumors, inducing superior anti-tumor activity while reducing systemic T cell dysfunction and promoting memory formation. Persisting peptide/IFA vaccine depots can induce specific T cell sequestration, dysfunction and deletion at vaccination sites; short-lived formulations may overcome these limitations and result in greater therapeutic efficacy of peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Yared Hailemichael
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhu Z, Singh V, Watkins SK, Bronte V, Shoe JL, Feigenbaum L, Hurwitz AA. High-avidity T cells are preferentially tolerized in the tumor microenvironment. Cancer Res 2012. [PMID: 23204239 DOI: 10.1158/0008-5472.can-12-1123] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
One obstacle in eliciting potent antitumor immune responses is the induction of tolerance to tumor antigens. TCR(lo) mice bearing a TCR transgene specific for the melanoma antigen tyrosinase-related protein-2 (TRP-2, Dct) harbor T cells that maintain tumor antigen responsiveness but lack the ability to control melanoma outgrowth. We used this model to determine whether higher avidity T cells could control tumor growth without becoming tolerized. As a part of the current study, we developed a second TRP-2-specific TCR transgenic mouse line (TCR(hi)) that bears higher avidity T cells and spontaneously developed autoimmune depigmentation. In contrast to TCR(lo) T cells, which were ignorant of tumor-derived antigen, TCR(hi) T cells initially delayed subcutaneous B16 melanoma tumor growth. However, persistence in the tumor microenvironment resulted in reduced IFN-γ production and CD107a (Lamp1) mobilization, hallmarks of T-cell tolerization. IFN-γ expression by TCR(hi) T cells was critical for upregulation of MHC-I on tumor cells and control of tumor growth. Blockade of PD-1 signals prevented T-cell tolerization and restored tumor immunity. Depletion of tumor-associated dendritic cells (TADC) reduced tolerization of TCR(hi) T cells and enhanced their antitumor activity. In addition, TADCs tolerized TCR(hi) T cells but not TCR(lo) T cells in vitro. Our findings show that T-cell avidity is a critical determinant of not only tumor control but also susceptibility to tolerization in the tumor microenvironment. For this reason, care should be exercised when considering T-cell avidity in designing cancer immunotherapeutics.
Collapse
Affiliation(s)
- Ziqiang Zhu
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, NCI, Frederick, MD 21702, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Öhman J, Magnusson B, Telemo E, Jontell M, Hasséus B. Langerhans cells and T cells sense cell dysplasia in oral leukoplakias and oral squamous cell carcinomas--evidence for immunosurveillance. Scand J Immunol 2012; 76:39-48. [PMID: 22469080 DOI: 10.1111/j.1365-3083.2012.02701.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Leukoplakias (LPLs) are lesions in the oral mucosa that may develop into oral squamous cell carcinoma (OSCC). The objective of this study was to assess presence and distribution of dendritic Langerhans cells (LCs) and T cells in patients with LPLs with or without cell dysplasia and in oral squamous cell carcinoma (OSCC). Biopsy specimens from patients with leukoplakias (LPLs) with or without dysplasia and oral squamous cell carcinoma (OSCC) were immunostained with antibodies against CD1a, Langerin, CD3, CD4, CD8 and Ki67, followed by quantitative analysis. Analyses of epithelium and connective tissue revealed a significantly higher number of CD1a + LCs in LPLs with dysplasia compared with LPLs without dysplasia. Presence of Langerin + LCs in epithelium did not differ significantly between LPLs either with or without dysplasia and OSCC. T cells were found in significantly increased numbers in LPLs with dysplasia and OSCC. The number of CD4+ cells did not differ significantly between LPLs with and without dysplasia, but a significant increase was detected when comparing LPLs with dysplasia with OSCC. CD8+ cells were significantly more abundant in OSCC and LPLs with dysplasia compared with LPLs without dysplasia. Proliferating cells (Ki67+) were significantly more abundant in OSCC compared to LPLs with dysplasia. Confocal laser scanning microscopy revealed colocalization of LCs and T cells in LPLs with dysplasia and in OSCC. LCs and T cells are more numerous in tissue compartments with dysplastic epithelial cells and dramatically increase in OSCC. This indicates an ongoing immune response against cells with dysplasia.
Collapse
Affiliation(s)
- J Öhman
- Department of Oral Medicine and Pathology, Institute of Odontology, Gothenburg, Sweden.
| | | | | | | | | |
Collapse
|
37
|
Kaluza KM, Kottke T, Diaz RM, Rommelfanger D, Thompson J, Vile R. Adoptive transfer of cytotoxic T lymphocytes targeting two different antigens limits antigen loss and tumor escape. Hum Gene Ther 2012; 23:1054-64. [PMID: 22734672 DOI: 10.1089/hum.2012.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
An antitumor T-cell response can lead to tumor control without clearing all tumor cells. As long as residual tumor cells remain, there is a constant risk of escape from that T-cell response. We previously showed that adoptive transfer of anti-ova OT-I T cells into B16ova-bearing mice led to tumor regression followed by escape of tumors that had lost the ova gene, rendering the OT-I T cells ineffective. In this study, we hypothesized that simultaneous transfer of cytotoxic T lymphocytes targeted against two independent antigens would reduce selection for single-antigen-loss cells, thereby limiting tumor escape. Using OT-I and Pmel T cells to treat B16ova tumors, we found that early cotransfer could prevent tumor emergence in most mice, whereas neither T-cell specificity alone was able to do so. When combined with total body irradiation for the treatment of larger 7-day tumors, cotransfer was also better at limiting tumor recurrence, and the tumors that did escape combination therapy continued to express both target antigens. As adoptively transferred T cells also persisted in vivo, even in mice with recurrent tumors, we hypothesized that restimulation of these antitumor T cells would prolong survival of mice with recurrent tumors. Consistent with this hypothesis, administration of a low-dose regimen of cyclophosphamide following tumor escape slowed tumor growth in mice that had previously received T-cell therapy, but not in control-treated mice, an effect that was associated with increased activation of T cells in vitro by low- but not high-dose cyclophosphamide.
Collapse
Affiliation(s)
- Karen M Kaluza
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
38
|
Prinz PU, Mendler AN, Masouris I, Durner L, Oberneder R, Noessner E. High DGK-α and Disabled MAPK Pathways Cause Dysfunction of Human Tumor-Infiltrating CD8+ T Cells That Is Reversible by Pharmacologic Intervention. THE JOURNAL OF IMMUNOLOGY 2012; 188:5990-6000. [DOI: 10.4049/jimmunol.1103028] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
39
|
Weiss VL, Lee TH, Song H, Kouo TS, Black CM, Sgouros G, Jaffee EM, Armstrong TD. Trafficking of high avidity HER-2/neu-specific T cells into HER-2/neu-expressing tumors after depletion of effector/memory-like regulatory T cells. PLoS One 2012; 7:e31962. [PMID: 22359647 PMCID: PMC3281086 DOI: 10.1371/journal.pone.0031962] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/16/2012] [Indexed: 02/02/2023] Open
Abstract
Background Cancer vaccines are designed to activate and enhance cancer-antigen-targeted T cells that are suppressed through multiple mechanisms of immune tolerance in cancer-bearing hosts. T regulatory cell (Treg) suppression of tumor-specific T cells is one barrier to effective immunization. A second mechanism is the deletion of high avidity tumor-specific T cells, which leaves a less effective low avidity tumor specific T cell repertoire available for activation by vaccines. Treg depleting agents including low dose cyclophosphamide (Cy) and antibodies that deplete CD25-expressing Tregs have been used with limited success to enhance the potency of tumor-specific vaccines. In addition, few studies have evaluated mechanisms that activate low avidity cancer antigen-specific T cells. Therefore, we developed high and low avidity HER-2/neu-specific TCR transgenic mouse colonies specific for the same HER-2/neu epitope to define the tolerance mechanisms that specifically affect high versus low avidity tumor-specific T cells. Methodology/Principal Findings High and low avidity CD8+ T cell receptor (TCR) transgenic mice specific for the breast cancer antigen HER-2/neu (neu) were developed to provide a purified source of naïve, tumor-specific T cells that can be used to study tolerance mechanisms. Adoptive transfer studies into tolerant FVB/N-derived HER-2/neu transgenic (neu-N) mice demonstrated that high avidity, but not low avidity, neu-specific T cells are inhibited by Tregs as the dominant tolerizing mechanism. High avidity T cells persisted, produced IFNγ, trafficked into tumors, and lysed tumors after adoptive transfer into mice treated with a neu-specific vaccine and low dose Cy to deplete Tregs. Analysis of Treg subsets revealed a Cy-sensitive CD4+Foxp3+CD25low tumor-seeking migratory phenotype, characteristic of effector/memory Tregs, and capable of high avidity T cell suppression. Conclusion/Significance Depletion of CD25low Tregs allows activation of tumor-clearing high avidity T cells. Thus, the development of agents that specifically deplete Treg subsets should translate into more effective immunotherapies while avoiding autoimmunity.
Collapse
Affiliation(s)
- Vivian L. Weiss
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Immunology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Timothy H. Lee
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hong Song
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Radiology and Nuclear Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Theodore S. Kouo
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Immunology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chelsea M. Black
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Cellular and Molecular Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - George Sgouros
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Radiology and Nuclear Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Elizabeth M. Jaffee
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Immunology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Cellular and Molecular Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Pharmacology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Skip Viragh Pancreatic Cancer Center, and Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Todd D. Armstrong
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
Tatari-Calderone Z, Stojakovic M, Dewan R, Le Bouder G, Jankovic D, Vukmanovic S. Age-related accumulation of T cells with markers of relatively stronger autoreactivity leads to functional erosion of T cells. BMC Immunol 2012; 13:8. [PMID: 22321827 PMCID: PMC3305419 DOI: 10.1186/1471-2172-13-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 02/09/2012] [Indexed: 11/10/2022] Open
Abstract
Background Thymic involution is a prominent characteristic of an aging immune system. When thymic function is reduced/absent, the peripheral T cell pool is subject to the laws of peripheral T cell homeostasis that favor survival/expansion of T cell receptors with relatively higher functional avidity for self-peptide/MHC complexes. Due to difficulties in assessing the TCR avidity in polyclonal population of T cells, it is currently not known whether high avidity T cells preferentially survive in aging individuals, and what impact this might have on the function of the immune system and development of autoimmune diseases. Results The phenotype of T cells from aged mice (18-24 months) indicating functional TCR avidity (CD3 and CD5 expression) correlates with the level of preserved thymic function. In mice with moderate thymic output (> 30% of peripheral CD62Lhi T cells), T cells displayed CD3lowCD5hi phenotype characteristic for high functional avidity. In old mice with drastically low numbers of CD62Lhi T cells reduced CD5 levels were found. After adult thymectomy, T cells of young mice developed CD3lowCD5hi phenotype, followed by a CD3lowCD5low phenotype. Spleens of old mice with the CD3low/CD5hi T cell phenotype displayed increased levels of IL-10 mRNA, and their T cells could be induced to secrete IL-10 in vitro. In contrast, downmodulation of CD5 was accompanied with reduced IL-10 expression and impaired anti-CD3 induced proliferation. Irrespective of the CD3/CD5 phenotype, reduced severity of experimental allergic myelitis occurred in old mice. In MTB TCRβ transgenic mice that display globally elevated TCR avidity for self peptide/MHC, identical change patterns occurred, only at an accelerated pace. Conclusions These findings suggest that age-associated dysfunctions of the immune system could in part be due to functional erosion of T cells devised to protect the hosts from the prolonged exposure to T cells with high-avidity for self.
Collapse
Affiliation(s)
- Zohreh Tatari-Calderone
- Center for Cancer and Immunology Research, Children's Research Institute, Washington, DC, USA
| | | | | | | | | | | |
Collapse
|
41
|
Mendler AN, Hu B, Prinz PU, Kreutz M, Gottfried E, Noessner E. Tumor lactic acidosis suppresses CTL function by inhibition of p38 and JNK/c-Jun activation. Int J Cancer 2011; 131:633-40. [DOI: 10.1002/ijc.26410] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 08/11/2011] [Indexed: 12/20/2022]
|
42
|
Hotta K, Sho M, Fujimoto K, Shimada K, Yamato I, Anai S, Konishi N, Hirao Y, Nonomura K, Nakajima Y. Prognostic significance of CD45RO+ memory T cells in renal cell carcinoma. Br J Cancer 2011; 105:1191-6. [PMID: 21934683 PMCID: PMC3208496 DOI: 10.1038/bjc.2011.368] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background: Memory T cells are well known to have a critical role for host defense in humans. However, their role in actual human cancer remains largely unknown. In this study, we tried to reveal the clinical importance of tumour-infiltrating CD45RO+ memory T cells in renal cell carcinoma (RCC). Methods: We analysed 105 patients with RCC, who received radical or partial nephrectomy. Those were 65 in TNM stage I, 7 in stage II, 15 in stage III, and 18 in stage IV, respectively. CD45RO expression was evaluated by immunohistochemistry. CD4 and CD8 expressions were also systematically assessed in the same manner. Results: Patients with higher TNM stage or high nuclear grade were found to have higher densities of CD45RO. Furthermore, CD45RO status was positively correlated with preoperative C-reactive protein level. In prognostic analysis, CD45RO+lo patients had a significantly better prognosis than CD45RO+hi patients. There was also a significant difference between CD4+lo and CD4+hi groups, whereas no significant difference was observed in CD8 T-cell status. Finally, multivariate analysis revealed that CD45RO+ status was the independent prognostic factor for patient overall survival. Conclusion: CD45RO+ memory T-cell status has a significant independent prognostic value, indicating that the adaptive immune response is functionally critical in human RCC.
Collapse
Affiliation(s)
- K Hotta
- Department of Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Krishnadas DK, Stamer MM, Dunham K, Bao L, Lucas KG. Wilms' tumor 1-specific cytotoxic T lymphocytes can be expanded from adult donors and cord blood. Leuk Res 2011; 35:1520-6. [PMID: 21774984 DOI: 10.1016/j.leukres.2011.06.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 05/12/2011] [Accepted: 06/28/2011] [Indexed: 01/01/2023]
Abstract
The use of WT1-specific CTL is one potential strategy to treat leukemic relapse following allogeneic stem cell transplant (SCT). Previous studies have largely focused on generating WT1-CTL from adult donors by cloning. We demonstrate that WT1-CTL can be generated from healthy adult donors and from cord blood by stimulating with an overlapping pool of peptides derived from full length WT1 and selecting antigen-specific cells based on the expression of CD137. The rapid expansion with anti-CD3 and IL-2 resulted in a 100-200-fold expansion. These CTL lysed WT1 expressing targets, including leukemia lines, in a HLA restricted manner.
Collapse
Affiliation(s)
- Deepa K Krishnadas
- Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, Penn State College of Medicine, Hershey, PA 17033, USA.
| | | | | | | | | |
Collapse
|
44
|
Figel AM, Brech D, Prinz PU, Lettenmeyer UK, Eckl J, Turqueti-Neves A, Mysliwietz J, Anz D, Rieth N, Muenchmeier N, Buchner A, Porubsky S, Siegert SI, Segerer S, Nelson PJ, Noessner E. Human renal cell carcinoma induces a dendritic cell subset that uses T-cell crosstalk for tumor-permissive milieu alterations. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:436-51. [PMID: 21703422 DOI: 10.1016/j.ajpath.2011.03.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/17/2011] [Accepted: 03/21/2011] [Indexed: 12/20/2022]
Abstract
Tissue dendritic cells (DCs) may influence the progression of renal cell carcinoma (RCC) by regulating the functional capacity of antitumor effector cells. DCs and their interaction with T cells were analyzed in human RCC and control kidney tissues. The frequency of CD209(+) DCs in RCCs was found to be associated with an unfavorable T(H)1 cell balance in the tissue and advanced tumor stages. The CD209(+) DCs in RCC were unusual because most of them co-expressed macrophage markers (CD14, CD163). The phenotype of these enriched-in-renal-carcinoma DCs (ercDCs) could be reiterated in vitro by carcinoma-secreted factors (CXCL8/IL-8, IL-6, and vascular endothelial growth factor). ErcDCs resembled conventional DCs in costimulatory molecule expression and antigen cross-presentation. They did not suppress cognate cytotoxic T-lymphocyte function and did not cause CD3ζ down-regulation, FOXP3 induction, or T-cell apoptosis in situ or in vitro; thus, they are different from classic myeloid-derived suppressor cells. ErcDCs secreted high levels of metalloproteinase 9 and used T-cell crosstalk to increase tumor-promoting tumor necrosis factor α and reduce chemokines relevant for T(H)1-polarized lymphocyte recruitment. This modulation of the tumor environment exerted by ercDCs suggests an immunologic mechanism by which tumor control can fail without involving cytotoxic T-lymphocyte inhibition. Pharmacologic targeting of the deviated DC differentiation could improve the efficacy of immunotherapy against RCC.
Collapse
Affiliation(s)
- Ainhoa-M Figel
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Song S, Zhang K, You H, Wang J, Wang Z, Yan C, Liu F. Significant anti-tumour activity of adoptively transferred T cells elicited by intratumoral dendritic cell vaccine injection through enhancing the ratio of CD8(+) T cell/regulatory T cells in tumour. Clin Exp Immunol 2010; 162:75-83. [PMID: 20735440 DOI: 10.1111/j.1365-2249.2010.04226.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have shown that immunization with dendritic cells (DCs) pulsed with hepatitis B virus core antigen virus-like particles (HBc-VLP) packaging with cytosine-guanine dinucleotide (CpG) (HBc-VLP/CpG) alone were able to delay melanoma growth but not able to eradicate the established tumour in mice. We tested whether, by modulating the vaccination approaches and injection times, the anti-tumour activity could be enhanced. We used a B16-HBc melanoma murine model not only to compare the efficacy of DC vaccine immunized via footpads, intravenously or via intratumoral injections in treating melanoma and priming tumour-specific immune responses, but also to observe how DC vaccination could improve the efficacy of adoptively transferred T cells to induce an enhanced anti-tumour immune response. Our results indicate that, although all vaccination approaches were able to protect mice from developing melanoma, only three intratumoral injections of DCs could induce a significant anti-tumour response. Furthermore, the combination of intratumoral DC vaccination and adoptive T cell transfer led to a more robust anti-tumour response than the use of each treatment individually by increasing CD8(+) T cells or the ratio of CD8(+) T cell/regulatory T cells in the tumour site. Moreover, the combination vaccination induced tumour-specific immune responses that led to tumour regression and protected surviving mice from tumour rechallenge, which is attributed to an increase in CD127-expressing and interferon-γ-producing CD8(+) T cells. Taken together, these results indicate that repeated intratumoral DC vaccination not only induces expansion of antigen-specific T cells against tumour-associated antigens in tumour sites, but also leads to elimination of pre-established tumours, supporting this combined approach as a potent strategy for DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- S Song
- Department of Molecular Biology and Key Laboratory of Experimental Animal, Hebei Medical University, Medical experimental Center, People's Hospital of Hebei Province, Shijiazhuang, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Tumor-resident CD8+ T-cell: the critical catalyst in IL-12-mediated reversal of tumor immune suppression. Arch Immunol Ther Exp (Warsz) 2010; 58:399-405. [PMID: 20872283 DOI: 10.1007/s00005-010-0097-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 06/21/2010] [Indexed: 01/29/2023]
Abstract
Tumor-resident T cells display a functionally impaired effector/memory (Tem) phenotype. Sustained intratumoral administration of IL-12, on the other hand, can restore cytolytic function to pre-existing CD8+ Tem, resulting in effective tumor kill. Whereas cytotoxic T lymphocytes (CTL) are generally assumed to mediate tumor regression via direct tumor cytotoxicity, recent work revealed that activated CD8+ Tem mobilize a systemic, multi-component effector cascade that includes both innate and adaptive immune mechanisms. Here we summarize these mechanisms, review how tumor-resident CD8+ Tem orchestrate this cascade and discuss the potential clinical implications of these findings.
Collapse
|
47
|
McDonnell AM, Prosser AC, van Bruggen I, Robinson BWS, Currie AJ. CD8alpha+ DC are not the sole subset cross-presenting cell-associated tumor antigens from a solid tumor. Eur J Immunol 2010; 40:1617-27. [PMID: 20373290 DOI: 10.1002/eji.200940153] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
One of the clear paradoxes in tumor immunology is the fact that cross-presentation of cell-associated tumor antigens to CD8(+) T cells is efficient, yet CTL generation is weak, and tumors continue to grow. We examined, for the first time whether this may be due to alterations in the phenotype or function of cross-presenting DC using a solid tumor model expressing a membrane bound neo-antigen (hemagglutinin, HA). Tumor antigen was constitutively cross-presented in the tumor-draining LN throughout tumor progression by CD11c(+) DC. Further analysis revealed that both CD8alpha(+) and CD8alpha(-) DC subsets, but not plasmacytoid DC, were effective at cross-presenting HA tumor antigen. The proportions of DC subsets in the tumor-draining LN were equivalent to those seen in the LN of naïve mice; however, a significant increase in the expression of the potential inhibitory B7 molecule, B7-DC, was noted and appeared to be restricted to the CD8alpha(-) DC subset. Therefore LN resident CD8alpha(+) DC are not the sole DC subset capable of cross-presenting cell-associated tumor antigens. Migratory tumor DC subsets with altered co-stimulatory receptor expression may contribute to induction and regulation of tumor-specific responses.
Collapse
Affiliation(s)
- Alison M McDonnell
- School of Medicine and Pharmacology, The University of Western Australia, Perth, Australia
| | | | | | | | | |
Collapse
|
48
|
Brayer J, Cheng F, Wang H, Horna P, Vicente-Suarez I, Pinilla-Ibarz J, Sotomayor EM. Enhanced CD8 T cell cross-presentation by macrophages with targeted disruption of STAT3. Immunol Lett 2010; 131:126-30. [PMID: 20346983 DOI: 10.1016/j.imlet.2010.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2009] [Revised: 02/11/2010] [Accepted: 03/19/2010] [Indexed: 11/28/2022]
Abstract
CD8 T cell tolerance, once thought to be largely a result of clonal deletion, is now appreciated to be much more complex, additionally involving multiple permutations of partial loss of effector function in residual clonal populations. This is especially important in the context of tumor immunity, in which persistent tolerized cytotoxic CD8 T cells (CTL), if reactivated, could potentially mount a protective response. Previously we have shown that antigen-presenting cells (APCs) with a targeted disruption of STAT3 break tolerance in CD4 T cells. Here we evaluate the STAT3-defective APC in terms of its ability to induce a productive CTL response. Our data demonstrate that macrophages derived from conditional STAT3 knockout mice are superior to wild-type macrophages in terms of their ability to prime cognate CTL responses, and to cross-present tumor-derived antigen to CTLs in vitro. CTLs cultured with STAT3-deficient APCs demonstrated a stronger proliferative response and produced increased amounts of IFN-gamma and TNF-alpha, all of which have been shown to be diminished in tumor-tolerized CD8 T cells. Targeting STAT3 signaling represents therefore an enticing strategy to augment CTL responses in the tumor-bearing host.
Collapse
Affiliation(s)
- Jason Brayer
- Department of Immunology and Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, United States
| | | | | | | | | | | | | |
Collapse
|
49
|
Sioud M. Does our current understanding of immune tolerance, autoimmunity, and immunosuppressive mechanisms facilitate the design of efficient cancer vaccines? Scand J Immunol 2009; 70:516-25. [PMID: 19906192 DOI: 10.1111/j.1365-3083.2009.02326.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The therapeutic use of the immune system to attack cancer cells has been a longstanding vision among tumour immunologists. However, most human tumours are poorly immunogenic and are able to invade the host immune system. Although these obstacles are clearly critical to cancer vaccine development, the induction of a strong anti-tumour immune response may rely on the activation of high affinity T cells through a molecular mimicry mechanism which involves cross-reactive recognition of foreign antigens mimicking the structure of tumour proteins. Taking into account the disparity in HLA molecules needed to present shared antigens; in late 1990s Stauss et al. described the possibility of generating allorestricted high affinity cytotoxic T cells against synthetic self-peptides bound to non-self-MHC molecules. In addition to the strategies indicated above, the inhibition of the immunosuppressive mechanisms associated with tumour invasion of the immune system using RNA interference also offers a new approach to vaccine design. This review highlights the problem of immune tolerance, the induction of autoreactive T cells, and describes strategies to enhance tumour immunity.
Collapse
Affiliation(s)
- M Sioud
- Department of Immunology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo, Norway.
| |
Collapse
|
50
|
Kasper HU, Drebber U, Stippel DL, Dienes HP, Gillessen A. Liver tumor infiltrating lymphocytes: Comparison of hepatocellular and cholangiolar carcinoma. World J Gastroenterol 2009; 15:5053-7. [PMID: 19859998 PMCID: PMC2768884 DOI: 10.3748/wjg.15.5053] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of tumor infiltrating lymphocytes (TIL) in primary hepatocellular and cholangiolar carcinomas of the liver.
METHODS: Immunohistochemical analysis was performed including antibodies to CD3, CD4, CD8, CD20, CD56 and TIA-1 in formalin-fixed and paraffin-embedded tissue of 35 liver resection specimens of hepatocellular or cholangiocellular carcinomas. Semiquantitative evaluation was performed with emphasis on the area of the tumor itself and of the tumor/liver interface.
RESULTS: All hepatocellular carcinomas showed infiltration of lymphocytes predominantly around the tumor in the tumor/liver interface consisting mainly of CD3+ CD4+ T lymphocytes [164.3/10 high power fields (HPF)] and in the tumor itself of CD8+ cells (54.9/10 HPF). Cholangiocarcinomas contained a heterogeneous amount of TIL, composed mainly of CD3+ T cells with a predominance of CD8+ cells in the tumor tissue (52.6/10 HPF) and of CD4+ cells in the interface region (223.1/10 HPF). CD56+ cells of the innate immune system were scarce. There was no significant difference between hepatocellular or cholangiolar carcinoma. No correlation with the clinicopathological data was seen.
CONCLUSION: Liver TIL consists of intratumoral CD8+ T cells and peritumoral CD4+ T cells independent of histogenetic origin. Different functions of lymphocytes in these regions seem possible.
Collapse
|