1
|
Bhatta M, Shenoy GN, Loyall JL, Gray BD, Bapardekar M, Conway A, Minderman H, Kelleher RJ, Carreno BM, Linette G, Shultz LD, Odunsi K, Balu-Iyer SV, Pak KY, Bankert RB. Novel phosphatidylserine-binding molecule enhances antitumor T-cell responses by targeting immunosuppressive exosomes in human tumor microenvironments. J Immunother Cancer 2021; 9:jitc-2021-003148. [PMID: 34599030 PMCID: PMC8488709 DOI: 10.1136/jitc-2021-003148] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2021] [Indexed: 12/21/2022] Open
Abstract
Background The human tumor microenvironment (TME) is a complex and dynamic milieu of diverse acellular and cellular components, creating an immunosuppressive environment, which contributes to tumor progression. We have previously shown that phosphatidylserine (PS) expressed on the surface of exosomes isolated from human TMEs is causally linked to T-cell immunosuppression, representing a potential immunotherapeutic target. In this study, we investigated the effect of ExoBlock, a novel PS-binding molecule, on T-cell responses in the TME. Methods We designed and synthesized a new compound, (ZnDPA)6-DP-15K, a multivalent PS binder named ExoBlock. The PS-binding avidity of ExoBlock was tested using an in vitro competition assay. The ability of this molecule to reverse exosome-mediated immunosuppression in vitro was tested using human T-cell activation assays. The in vivo therapeutic efficacy of ExoBlock was then tested in two different human tumor xenograft models, the melanoma-based xenomimetic (X-)mouse model, and the ovarian tumor-based omental tumor xenograft (OTX) model. Results ExoBlock was able to bind PS with high avidity and was found to consistently and significantly block the immunosuppressive activity of human ovarian tumor and melanoma-associated exosomes in vitro. ExoBlock was also able to significantly enhance T cell-mediated tumor suppression in vivo in both the X-mouse and the OTX model. In the X-mouse model, ExoBlock suppressed tumor recurrence in a T cell-dependent manner. In the OTX model, ExoBlock treatment resulted in an increase in the number as well as function of CD4 and CD8 T cells in the TME, which was associated with a reduction in tumor burden and metastasis, as well as in the number of circulating PS+ exosomes in tumor-bearing mice. Conclusion Our results establish that targeting exosomal PS in TMEs with ExoBlock represents a promising strategy to enhance antitumor T-cell responses.
Collapse
Affiliation(s)
| | - Gautam N Shenoy
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, USA
| | - Jenni L Loyall
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, USA
| | - Brian D Gray
- Molecular Targeting Technologies Inc, West Chester, Pennsylvania, USA
| | - Meghana Bapardekar
- Flow & Image Cytometry Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Alexis Conway
- Flow & Image Cytometry Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Hans Minderman
- Flow & Image Cytometry Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Raymond J Kelleher
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, USA
| | - Beatriz M Carreno
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Gerald Linette
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Kunle Odunsi
- University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo-The State University of New York, Buffalo, New York, USA
| | - Koon Yan Pak
- Molecular Targeting Technologies Inc, West Chester, Pennsylvania, USA
| | - Richard B Bankert
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
2
|
Shenoy GN, Bhatta M, Loyall JL, Kelleher RJ, Bernstein JM, Bankert RB. Exosomes Represent an Immune Suppressive T Cell Checkpoint in Human Chronic Inflammatory Microenvironments. Immunol Invest 2020; 49:726-743. [PMID: 32299258 PMCID: PMC7554261 DOI: 10.1080/08820139.2020.1748047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: T cells present in chronic inflammatory tissues such as nasal polyps (from chronic rhinosinusitis patients) have been demonstrated to be hypo-responsive to activation via the TCR, similar to tumor-specific T cells in multiple different human tumor microenvironments. While immunosuppressive exosomes have been known to contribute to the failure of the tumor-associated T cells to respond optimally to activation stimuli, it is not known whether they play a similar role in chronic inflammatory microenvironments. In the current study, we investigate whether exosomes derived from chronic inflammatory microenvironments contribute to the immune suppression of T cells. Methods: Exosomes were isolated by ultracentrifugation and characterized by size and composition using nanoparticle tracking analysis, scanning electron microscopy, antibody arrays and flow exometry. Immunosuppressive ability of the exosomes was measured by quantifying its effect on activation of T cells, using nuclear translocation of NFκB as an activation endpoint. Results: Exosomes were isolated and characterized from two different types of chronic inflammatory tissues - nasal polyps from chronic rhinosinusitis patients and synovial fluid from rheumatoid arthritis patients. These exosomes arrest the activation of T cells stimulated via the TCR. This immune suppression, like that which is seen in tumor microenvironments, is dependent in part upon a lipid, ganglioside GD3, which is expressed on the exosomal surface. Conclusion: Immunosuppressive exosomes present in non-malignant chronic inflammatory tissues represent a new T cell checkpoint, and potentially represent a novel therapeutic target to enhance the response to current therapies and prevent disease recurrences.
Collapse
Affiliation(s)
- Gautam N Shenoy
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | | | - Jenni L Loyall
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Raymond J Kelleher
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Joel M Bernstein
- Department of Otolaryngology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Richard B Bankert
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| |
Collapse
|
3
|
Luo H, Wu X, Sun R, Su J, Wang Y, Dong Y, Shi B, Sun Y, Jiang H, Li Z. Target-Dependent Expression of IL12 by synNotch Receptor-Engineered NK92 Cells Increases the Antitumor Activities of CAR-T Cells. Front Oncol 2019; 9:1448. [PMID: 31921693 PMCID: PMC6930917 DOI: 10.3389/fonc.2019.01448] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/04/2019] [Indexed: 01/04/2023] Open
Abstract
IL12 is an immune-stimulatory cytokine for key immune cells including T cells and NK cells. However, systemic administration of IL12 has serious side effects that limit its clinical application in patients. Recently, synthetic Notch (synNotch) receptors have been developed that induce transcriptional activation and deliver therapeutic payloads in response to the reorganization of specific antigens. NK92 cell is a human natural killer (NK) cell line which has been developed as tools for adjuvant immunotherapy of cancer. Here, we explored the possibility of using synNotch receptor-engineered NK92 cells to selectively secrete IL12 at the tumor site and increase the antitumor activities of chimeric antigen receptor (CAR)-modified T cells. Compared with the nuclear factor of activated T-cells (NFATs) responsive promoter, which is another regulatory element, the synNotch receptor was better at controlling the expression of cytokines. NK92 cells transduced with the GPC3-specific synNotch receptor could produce the proinflammatory cytokine IL12 (GPC3-Syn-IL12-NK92) in response to GPC3 antigen expressed in cancer cells. In vivo GPC3-Syn-IL12-NK92 cells controlling IL12 production could enhance the antitumor ability of GPC3-redirected CAR T cells and increase the infiltration of T cells without inducing toxicity. Taken together, our results demonstrated that IL12 supplementation by synNotch-engineered NK92 cells could secrete IL12 in a target-dependent manner, and promote the antitumor efficiency of CAR-T cells. Local expression of IL12 by synNotch-engineered NK92 cells might be a safe approach to enhance the clinical outcome of CAR-T cell therapy.
Collapse
Affiliation(s)
- Hong Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuqi Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingwen Su
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yiwei Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yansha Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,CARsgen Therapeutics, Shanghai, China
| |
Collapse
|
4
|
Shenoy GN, Loyall J, Berenson CS, Kelleher RJ, Iyer V, Balu-Iyer SV, Odunsi K, Bankert RB. Sialic Acid-Dependent Inhibition of T Cells by Exosomal Ganglioside GD3 in Ovarian Tumor Microenvironments. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:3750-3758. [PMID: 30446565 PMCID: PMC6289713 DOI: 10.4049/jimmunol.1801041] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/15/2018] [Indexed: 01/07/2023]
Abstract
The tumor microenvironment is rendered immunosuppressive by a variety of cellular and acellular factors that represent potential cancer therapeutic targets. Although exosomes isolated from ovarian tumor ascites fluids have been previously reported to induce a rapid and reversible T cell arrest, the factors present on or within exosomes that contribute to immunosuppression have not been fully defined. In this study, we establish that GD3, a ganglioside expressed on the surface of exosomes isolated from human ovarian tumor ascites fluids, is causally linked to the functional arrest of T cells activated through their TCR. This arrest is inhibited by Ab blockade of exosomal GD3 or by the removal of GD3+ exosomes. Empty liposomes expressing GD3 on the surface also inhibit the activation of T cells, establishing that GD3 contributes to the functional arrest of T cells independent of factors present in exosomes. Finally, we demonstrate that the GD3-mediated arrest of the TCR activation is dependent upon sialic acid groups, because their enzymatic removal from exosomes or liposomes results in a loss of inhibitory capacity. Collectively, these data define GD3 as a potential immunotherapeutic target.
Collapse
Affiliation(s)
- Gautam N. Shenoy
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Jenni Loyall
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Charles S. Berenson
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, Infectious Disease Division, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, and Department of Veteran Affairs, Western New York Health Care System, Buffalo, New York
| | - Raymond J. Kelleher
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Vandana Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Sathy V. Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York
| | - Richard B. Bankert
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| |
Collapse
|
5
|
Kosti P, Maher J, Arnold JN. Perspectives on Chimeric Antigen Receptor T-Cell Immunotherapy for Solid Tumors. Front Immunol 2018; 9:1104. [PMID: 29872437 PMCID: PMC5972325 DOI: 10.3389/fimmu.2018.01104] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/02/2018] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy entails the genetic engineering of a patient's T-cells to express membrane spanning fusion receptors with defined specificities for tumor-associated antigens. These CARs are capable of eliciting robust T-cell activation to initiate killing of the target tumor cells. This therapeutic approach has produced unprecedented clinical outcomes in the treatment of "liquid" hematologic cancers, but to date has not produced comparable responses in targeting solid malignancies. Advances in our understanding of the immunobiology of solid tumors have highlighted several hurdles which currently hinder the efficacy of this therapy. These barriers include the insufficient accumulation of CAR T-cells in the tumor due to poor trafficking or physical exclusion and the exposure of infiltrating CAR T-cells to a panoply of immune suppressive checkpoint molecules, cytokines, and metabolic stresses that are not conducive to efficient immune reactions and can thereby render these cells anergic, exhausted, or apoptotic. This mini-review summarizes these hurdles and describes some recent approaches and innovations to genetically re-engineer CAR T-cells to counter inhibitory influences found in the tumor microenvironment. Novel immunotherapy drug combinations to potentiate the activity of CAR T-cells are also discussed. As our understanding of the immune landscape of tumors improves and our repertoire of immunotherapeutic drugs expands, it is envisaged that the efficacy of CAR T-cells against solid tumors might be potentiated using combination therapies, which it is hoped may lead to meaningful improvements in clinical outcome for patients with refractory solid malignancies.
Collapse
Affiliation(s)
- Paris Kosti
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - John Maher
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom.,Department of Immunology, Eastbourne Hospital, Eastbourne, East Sussex, United Kingdom.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - James N Arnold
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
6
|
Karsten MM, Enders S, Knabl J, Kirn V, Düwell P, Rack B, Blohmer JU, Mayr D, Dian D. Biologic meshes and synthetic meshes in cancer patients: a double-edged sword: differences in production of IL-6 and IL-12 caused by acellular dermal matrices in human immune cells. Arch Gynecol Obstet 2018; 297:1265-1270. [PMID: 29417284 DOI: 10.1007/s00404-018-4710-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 02/01/2018] [Indexed: 01/31/2023]
Abstract
PURPOSE In 2005, Breuing et al. first described the use of acellular dermal matrices (ADMs) in breast cancer patients. ADMs are assumed to be safe to use in an oncologic setting, but data from controlled studies are still needed. Here, we investigate the effects of ADMs on the production of interleukin (IL)-6 and IL-12, key regulators of immune suppression and activation. METHODS Strattice (ST), CollaMend (CM), and Biodesign (BD) biologic meshes and TiLoop, a synthetic mesh (TL), were used in this study. We isolated myeloid dendritic cells (MDCs), untouched plasmacytoid dendritic cells (pDCs), naïve B cells, and CD8+ T cells and co-cultured these cells with either the biologic meshes or TL. As positive controls, we used CpG ODN 2216 or lipopolysaccharide (LPS). The cytokine concentrations of IL-12p70 and IL-6 were determined after 7 days using sandwich ELISA sets. RESULTS There were highly significant differences between the ADMs and TL in terms of their ability to stimulate immunologic responses. IL-6 expression was significantly increased in B cells (p = 0.0006131) and T cells (p = 0.00418) when comparing TL and ADMs. We also identified significant differences in IL-12 production by B cells (p = 0.0166) and T cells (p = 0.003636) when comparing TL and ADMs. CONCLUSIONS Despite the assumed lack of an immunological response to ADMs, in our experimental study, human immune cells reacted with significantly different cytokine profiles. These findings may have implications for the potential activation or suppression of effector cells in cancer patients and could explain some of the post clinical post surgical signs of ADMS like skin rush and seroma.
Collapse
Affiliation(s)
- Maria Margarete Karsten
- Klinik für Gynäkologie und Brustzentrum, Charité Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117, Berlin, Germany.
| | - Sabine Enders
- Klinik für Frauenheilkunde, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julia Knabl
- Klinik für Frauenheilkunde, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Verena Kirn
- Klinik und Poliklinik für Frauenheilkunde, Universität Köln, Cologne, Germany
| | - Peter Düwell
- Abteilung für klinische Pharmakologie, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Brigitte Rack
- Klinik für Frauenheilkunde, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jens-Uwe Blohmer
- Klinik für Gynäkologie und Brustzentrum, Charité Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Doris Mayr
- Abteilung für Pathologie, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Darius Dian
- Klinik für Frauenheilkunde, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
7
|
Shenoy GN, Loyall J, Maguire O, Iyer V, Kelleher RJ, Minderman H, Wallace PK, Odunsi K, Balu-Iyer SV, Bankert RB. Exosomes Associated with Human Ovarian Tumors Harbor a Reversible Checkpoint of T-cell Responses. Cancer Immunol Res 2018; 6:236-247. [PMID: 29301753 DOI: 10.1158/2326-6066.cir-17-0113] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/09/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022]
Abstract
Nano-sized membrane-encapsulated extracellular vesicles isolated from the ascites fluids of ovarian cancer patients are identified as exosomes based on their biophysical and compositional characteristics. We report here that T cells pulsed with these tumor-associated exosomes during TCR-dependent activation inhibit various activation endpoints including translocation of NFκB and NFAT into the nucleus, upregulation of CD69 and CD107a, production of cytokines, and cell proliferation. In addition, the activation of virus-specific CD8+ T cells that are stimulated with the cognate viral peptides presented in the context of class I MHC is also suppressed by the exosomes. The inhibition occurs without loss of cell viability and coincidentally with the binding and internalization of these exosomes. This exosome-mediated inhibition of T cells was transient and reversible: T cells exposed to exosomes can be reactivated once exosomes are removed. We conclude that tumor-associated exosomes are immunosuppressive and represent a therapeutic target, blockade of which would enhance the antitumor response of quiescent tumor-associated T cells and prevent the functional arrest of adoptively transferred tumor-specific T cells or chimeric antigen receptor T cells. Cancer Immunol Res; 6(2); 236-47. ©2018 AACR.
Collapse
Affiliation(s)
- Gautam N Shenoy
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, New York
| | - Jenni Loyall
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, New York
| | - Orla Maguire
- Flow and Image Cytometry Shared Resource, Roswell Park Cancer Institute, Buffalo, New York
| | - Vandana Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Raymond J Kelleher
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, New York
| | - Hans Minderman
- Flow and Image Cytometry Shared Resource, Roswell Park Cancer Institute, Buffalo, New York
| | - Paul K Wallace
- Department of Flow Cytometry, Roswell Park Cancer Institute, Buffalo, New York
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Richard B Bankert
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, New York.
| |
Collapse
|
8
|
Davoodzadeh Gholami M, Kardar GA, Saeedi Y, Heydari S, Garssen J, Falak R. Exhaustion of T lymphocytes in the tumor microenvironment: Significance and effective mechanisms. Cell Immunol 2017; 322:1-14. [PMID: 29079339 DOI: 10.1016/j.cellimm.2017.10.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/23/2022]
Abstract
T lymphocytes play crucial roles in adaptive immune responses to tumors. However, due to different tolerance mechanisms and inhibitory effects of the tumor microenvironment (TME) on T cells, responses to tumors are insufficient. In fact, cellular and molecular suppressive mechanisms repress T cell responses in the TME, resulting in senescent, anergic and exhausted lymphocytes. Exhaustion is a poor responsive status of T cells, with up-regulated expression of inhibitory receptors, decreased production of effective cytokines, and reduced cytotoxic activity. Low immunogenicity of tumor antigens and inadequate presentation of tumor-specific antigens results in inappropriate activation of naive T lymphocytes against tumor antigens. Moreover, when effector cytotoxic T cells enter TME, they encounter a complicated network of cells and cytokines that suppress their effectiveness and turn them into exhausted T cells. Thus, the mechanism of T cell exhaustion in cancer is different from that in chronic infections. In this review we will discuss the main components such as inhibitory receptors, inflammatory cells, stromal cells, cytokine milieu as well as environmental and metabolic conditions in TME which play role in development of exhaustion. Furthermore, recent therapeutic methods available to overcome exhaustion will be discussed.
Collapse
Affiliation(s)
- Mohammad Davoodzadeh Gholami
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholam Ali Kardar
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Yousef Saeedi
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Sahel Heydari
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Scarfò I, Maus MV. Current approaches to increase CAR T cell potency in solid tumors: targeting the tumor microenvironment. J Immunother Cancer 2017; 5:28. [PMID: 28331617 PMCID: PMC5359946 DOI: 10.1186/s40425-017-0230-9] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 02/24/2017] [Indexed: 01/02/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a revolutionary treatment for haematological malignancies (i.e. B-ALL). However, the success of this type of treatment has not yet been achieved in solid tumors. One hypothesis is that the immunosuppressive nature of the tumor microenvironment (TME) influences and affects the efficacy of adoptive immunotherapy. Understanding the role of the TME and its interaction with CAR T-cells is crucial to improve the potency of adoptive immunotherapy. In this review, we discuss the strategies and potential combinatorial approaches recently developed in mouse models to enhance the efficacy of CAR T-cells, with particular emphasis on the translational potential of these approaches.
Collapse
Affiliation(s)
- Irene Scarfò
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, 149 13th Street, Room 7.219, Charlestown, Boston, MA 02129 USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, 149 13th Street, Room 7.219, Charlestown, Boston, MA 02129 USA
| |
Collapse
|
10
|
Evaristo C, Spranger S, Barnes SE, Miller ML, Molinero LL, Locke FL, Gajewski TF, Alegre ML. Cutting Edge: Engineering Active IKKβ in T Cells Drives Tumor Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:2933-8. [PMID: 26903482 PMCID: PMC4799771 DOI: 10.4049/jimmunol.1501144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 02/01/2016] [Indexed: 01/02/2023]
Abstract
Acquired dysfunction of tumor-reactive T cells is one mechanism by which tumors can evade the immune system. Identifying and correcting pathways that contribute to such dysfunction should enable novel anticancer therapy design. During cancer growth, T cells show reduced NF-κB activity, which is required for tumor rejection. Impaired T cell-intrinsic NF-κB may create a vicious cycle conducive to tumor progression and further T cell dysfunction. We hypothesized that forcing T cell-intrinsic NF-κB activation might break this cycle and induce tumor elimination. NF-κB was activated in T cells by inducing the expression of a constitutively active form of the upstream activator IκB kinase β (IKKβ). T cell-restricted constitutively active IKKβ augmented the frequency of functional tumor-specific CD8(+) T cells and improved tumor control. Transfer of constitutively active IKKβ-transduced T cells also boosted endogenous T cell responses that controlled pre-established tumors. Our results demonstrate that driving T cell-intrinsic NF-κB can result in tumor control, thus identifying a pathway with potential clinical applicability.
Collapse
Affiliation(s)
- César Evaristo
- Department of Medicine, University of Chicago, Chicago, IL 60637; and
| | - Stefani Spranger
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Sarah E Barnes
- Department of Medicine, University of Chicago, Chicago, IL 60637; and
| | - Michelle L Miller
- Department of Medicine, University of Chicago, Chicago, IL 60637; and
| | | | - Frederick L Locke
- Department of Medicine, University of Chicago, Chicago, IL 60637; and
| | - Thomas F Gajewski
- Department of Medicine, University of Chicago, Chicago, IL 60637; and Department of Pathology, University of Chicago, Chicago, IL 60637
| | | |
Collapse
|
11
|
Ma X, Yan W, Zheng H, Du Q, Zhang L, Ban Y, Li N, Wei F. Regulation of IL-10 and IL-12 production and function in macrophages and dendritic cells. F1000Res 2015; 4. [PMID: 26918147 PMCID: PMC4754024 DOI: 10.12688/f1000research.7010.1] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
Interleukin-10 and Interleukin-12 are produced primarily by pathogen-activated antigen-presenting cells, particularly macrophages and dendritic cells. IL-10 and IL-12 play very important immunoregulatory roles in host defense and immune homeostasis. Being anti- and pro-inflammatory in nature, respectively, their functions are antagonistically opposing. A comprehensive and in-depth understanding of their immunological properties and signaling mechanisms will help develop better clinical intervention strategies in therapy for a wide range of human disorders. Here, we provide an update on some emerging concepts, controversies, unanswered questions, and opinions regarding the immune signaling of IL-10 and IL-12.
Collapse
Affiliation(s)
- Xiaojing Ma
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Wenjun Yan
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Hua Zheng
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Qinglin Du
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Lixing Zhang
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Yi Ban
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Na Li
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Fang Wei
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| |
Collapse
|
12
|
Kelleher RJ, Balu-Iyer S, Loyall J, Sacca AJ, Shenoy GN, Peng P, Iyer V, Fathallah AM, Berenson CS, Wallace PK, Tario J, Odunsi K, Bankert RB. Extracellular Vesicles Present in Human Ovarian Tumor Microenvironments Induce a Phosphatidylserine-Dependent Arrest in the T-cell Signaling Cascade. Cancer Immunol Res 2015; 3:1269-78. [PMID: 26112921 DOI: 10.1158/2326-6066.cir-15-0086] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022]
Abstract
The identification of immunosuppressive factors within human tumor microenvironments, and the ability to block these factors, would be expected to enhance patients' antitumor immune responses. We previously established that an unidentified factor, or factors, present in ovarian tumor ascites fluids reversibly inhibited the activation of T cells by arresting the T-cell signaling cascade. Ultracentrifugation of the tumor ascites fluid has now revealed a pellet that contains small extracellular vesicles (EV) with an average diameter of 80 nm. The T-cell arrest was determined to be causally linked to phosphatidylserine (PS) that is present on the outer leaflet of the vesicle bilayer, as a depletion of PS-expressing EV or a blockade of PS with anti-PS antibody significantly inhibits the vesicle-induced signaling arrest. The inhibitory EV were also isolated from solid tumor tissues. The presence of immunosuppressive vesicles in the microenvironments of ovarian tumors and our ability to block their inhibition of T-cell function represent a potential therapeutic target for patients with ovarian cancer.
Collapse
Affiliation(s)
- Raymond J Kelleher
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York
| | - Sathy Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Jenni Loyall
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York
| | - Anthony J Sacca
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York
| | - Gautam N Shenoy
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York
| | - Peng Peng
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York
| | - Vandana Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Anas M Fathallah
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Charles S Berenson
- School of Medicine, Infectious Disease Division, University at Buffalo, Buffalo, New York, and Department of Veteran Affairs, Western New York Health Care System, Buffalo, New York
| | - Paul K Wallace
- Department of Flow Cytometry, Roswell Park Cancer Institute, Buffalo, New York
| | - Joseph Tario
- Department of Flow Cytometry, Roswell Park Cancer Institute, Buffalo, New York
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Richard B Bankert
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York.
| |
Collapse
|
13
|
Koneru M, O'Cearbhaill R, Pendharkar S, Spriggs DR, Brentjens RJ. A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J Transl Med 2015; 13:102. [PMID: 25890361 PMCID: PMC4438636 DOI: 10.1186/s12967-015-0460-x] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/11/2015] [Indexed: 12/15/2022] Open
Abstract
Purpose Recurrent platinum-resistant ovarian cancer has no curative options, necessitating the development of novel treatments, including immunotherapy. Rationale Patient-derived T cells can be genetically modified to express chimeric antigen receptors (CARs) specific to tumor-associated antigens in an HLA-independent manner, with promising preclinical results. MUC16ecto is highly expressed on most epithelial ovarian carcinomas but at low levels on normal tissues, offering an excellent immunotherapeutic target for this cancer. CAR T cells further modified to secrete IL-12 show enhanced cytotoxicity, persistence, and modulation of the tumor microenvironment. Design We propose a dose escalation phase I clinical trial for patients with recurrent MUC-16ecto+ ovarian cancer to test the safety of intravenous and intraperitoneal administration and the preliminary efficacy of autologous IL-12 secreting, MUC-16ecto CAR T cells containing a safety elimination gene. Innovation This trial targets MUC-16ecto, a novel and promising tumor-associated antigen. This will be the first time CAR T cells are injected intraperitoneally directly into the site of the tumor within the abdomen in humans. Furthermore, the ability of genetically modified cells to secrete IL-12 will potentially enhance CAR T cell persistence and modulate the tumor microenvironment. For safety purposes, an elimination gene has been incorporated into the CAR T cells to mitigate any on-target, off-tumor or other unforeseen toxicity.
Collapse
Affiliation(s)
- Mythili Koneru
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,Weill Cornell Medical College, New York, NY, USA.
| | - Roisin O'Cearbhaill
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,Weill Cornell Medical College, New York, NY, USA.
| | - Swati Pendharkar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - David R Spriggs
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,Weill Cornell Medical College, New York, NY, USA.
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
14
|
Koneru M, Purdon TJ, Spriggs D, Koneru S, Brentjens RJ. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology 2015; 4:e994446. [PMID: 25949921 DOI: 10.4161/2162402x.2014.994446] [Citation(s) in RCA: 351] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/28/2014] [Indexed: 02/07/2023] Open
Abstract
A novel approach for the treatment of ovarian cancer includes immunotherapy with genetically engineered T cells targeted to ovarian cancer cell antigens. Using retroviral transduction, T cells can be created that express an artificial T cell receptor (TCR) termed a chimeric antigen receptor (CAR). We have generated a CAR, 4H11-28z, specific to MUC-16ecto antigen, which is the over-expressed on a majority of ovarian tumor cells and is the retained portion of MUC-16 after cleavage of CA-125. We previously demonstrated that T cells modified to express the 4H11-28z CAR eradicate orthotopic human ovarian cancer xenografts in SCID-Beige mice. However, despite the ability of CAR T cells to localize to tumors, their activation in the clinical setting can be inhibited by the tumor microenvironment, as is commonly seen for endogenous antitumor immune response. To potentially overcome this limitation, we have recently developed a construct that co-expresses both MUC16ecto CAR and IL-12 (4H11-28z/IL-12). In vitro, 4H11-28z/IL-12 CAR T cells show enhanced proliferation and robust IFNγ secretion compared to 4H11-28z CAR T cells. In SCID-Beige mice with human ovarian cancer xenografts, IL-12 secreting CAR T cells exhibit enhanced antitumor efficacy as determined by increased survival, prolonged persistence of T cells, and higher systemic IFNγ. Furthermore, in anticipation of translating these results into a phase I clinical trial which will be the first to study IL-12 secreting CAR T cells in ovarian cancer, an elimination gene has been included to allow for deletion of CAR T cells in the context of unforeseen or off-tumor on-target toxicity.
Collapse
Key Words
- AAPCs, artificial antigen presenting cells; ADCC, antibody-dependent cellular cytotoxicity; ALL, acute lymphocytic leukemia; CAR, chimeric antigen receptor; EGFRt, truncated epidermal growth factor; EOC, epithelial ovarian cancer; i.p., intraperitoneal; IL-12, interleukin-12; i.v., intravenous; MDSC, myeloid-derived suppressor cells; PBL, peripheral blood leukocytes; PBMCs, peripheral blood mononuclear cells; scFv, single-chain fragment antibody; TAA, tumor-associated antigen; TCR, T cell receptor; TIL, tumor-infiltrating lymphocytes; Tregs, regulatory T cells.
- IL-12
- MUC16
- chimeric antigen receptors
- human ovarian cancer
- tumor microenvironment
Collapse
Affiliation(s)
- Mythili Koneru
- Department of Medicine; Memorial Sloan-Kettering Cancer Center ; New York, NY, USA
| | - Terence J Purdon
- Department of Medicine; Memorial Sloan-Kettering Cancer Center ; New York, NY, USA
| | - David Spriggs
- Department of Medicine; Memorial Sloan-Kettering Cancer Center ; New York, NY, USA
| | | | - Renier J Brentjens
- Department of Medicine; Memorial Sloan-Kettering Cancer Center ; New York, NY, USA
| |
Collapse
|
15
|
Barnes SE, Wang Y, Chen L, Molinero LL, Gajewski TF, Evaristo C, Alegre ML. T cell-NF-κB activation is required for tumor control in vivo. J Immunother Cancer 2015; 3:1. [PMID: 25648675 PMCID: PMC4308877 DOI: 10.1186/s40425-014-0045-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/03/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND T cells have the capacity to eliminate tumors but the signaling pathways by which they do so are incompletely understood. T cell priming requires activation of the transcription factors AP-1, NFAT and NF-κB downstream of the TCR, but whether activation of T cell-NF-κB in vivo is required for tumor control has not been addressed. In humans and mice with progressively growing tumors, the activity of T cell-intrinsic NF-κB is often reduced. However, it is not clear if this is causal for an inability to reject transformed cells, or if it is a consequence of tumor growth. T cell-NF-κB is important for T cell survival and effector differentiation and plays an important role in enabling T cells to reject cardiac and islet allografts, suggesting the possibility that it may also be required for tumor elimination. In this study, we tested whether normal T cell-NF-κB activation is necessary for the rejection of tumors whose growth is normally controlled by the immune system. METHODS Mice with genetically impaired T cell-NF-κB activity were subcutaneously injected with MC57-SIY tumor cells. Tumor growth was measured over time, and the anti-tumor immune response was evaluated using flow cytometry and cytokine detection assays. RESULTS Mice with impaired T cell-NF-κB activity were unable to reject tumors that were otherwise eliminated by wildtype mice, despite equal accumulation of tumor-reactive T cells. In addition, specific impairment of NF-κB signaling downstream of the TCR was sufficient to prevent tumor rejection. Tumor antigen-specific T cell-IFN-γ and TNF-α production, as well as cytotoxic ability, were all reduced in mice with impaired T cell-NF-κB, suggesting an important role for this transcription factor in the effector differentiation of tumor-specific effector T cells. CONCLUSIONS Our results have identified the NF-κB pathway as an important signaling axis in T cells, required for the elimination of growing tumors in vivo. Maintaining or enhancing T cell-NF-κB activity may be a promising avenue for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Sarah E Barnes
- />Department of Medicine, The University of Chicago, 924 E. 57th St. JFK-R312, Chicago, IL 60637 USA
| | - Ying Wang
- />Department of Medicine, The University of Chicago, 924 E. 57th St. JFK-R312, Chicago, IL 60637 USA
| | - Luqiu Chen
- />Department of Medicine, The University of Chicago, 924 E. 57th St. JFK-R312, Chicago, IL 60637 USA
| | - Luciana L Molinero
- />Genentech, Inc., 1 DNA Way MS: 245c, South San Francisco, CA 94080 USA
| | - Thomas F Gajewski
- />Department of Medicine, The University of Chicago, 924 E. 57th St. JFK-R312, Chicago, IL 60637 USA
- />Department of Pathology, The University of Chicago, 927 E. 57th St, Chicago, IL 60637 USA
| | - Cesar Evaristo
- />Department of Medicine, The University of Chicago, 924 E. 57th St. JFK-R312, Chicago, IL 60637 USA
| | - Maria-Luisa Alegre
- />Department of Medicine, The University of Chicago, 924 E. 57th St. JFK-R312, Chicago, IL 60637 USA
| |
Collapse
|
16
|
Sheng L, Wang L, Sang X, Zhao X, Hong J, Cheng S, Yu X, Liu D, Xu B, Hu R, Sun Q, Cheng J, Cheng Z, Gui S, Hong F. Nano-sized titanium dioxide-induced splenic toxicity: a biological pathway explored using microarray technology. JOURNAL OF HAZARDOUS MATERIALS 2014; 278:180-188. [PMID: 24968254 DOI: 10.1016/j.jhazmat.2014.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 06/01/2014] [Accepted: 06/05/2014] [Indexed: 06/03/2023]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) have been widely used in various areas, and its potential toxicity has gained wide attention. However, the molecular mechanisms of multiple genes working together in the TiO2 NP-induced splenic injury are not well understood. In the present study, 2.5, 5, or 10mg/kg body weight TiO2 NPs were administered to the mice by intragastric administration for 90 consecutive days, their immune capacity in the spleen as well as the gene-expressed characteristics in the mouse damaged spleen were investigated using microarray assay. The findings showed that with increased dose, TiO2 NP exposure resulted in the increases of spleen indices, immune dysfunction, and severe macrophage infiltration as well as apoptosis in the spleen. Importantly, microarray data showed significant alterations in the expressions of 1041 genes involved in immune/inflammatory responses, apoptosis, oxidative stress, stress responses, metabolic processes, ion transport, signal transduction, cell proliferation/division, cytoskeleton and translation in the 10 mg/kg TiO2 NP-exposed spleen. Specifically, Cyp2e1, Sod3, Mt1, Mt2, Atf4, Chac1, H2-k1, Cxcl13, Ccl24, Cd14, Lbp, Cd80, Cd86, Cd28, Il7r, Il12a, Cfd, and Fcnb may be potential biomarkers of spleen toxicity following exposure to TiO2 NPs.
Collapse
Affiliation(s)
- Lei Sheng
- Medical College of Soochow University, Suzhou 215123, China
| | - Ling Wang
- Library of Soochow University, Suzhou 215123, China
| | - Xuezi Sang
- Medical College of Soochow University, Suzhou 215123, China
| | - Xiaoyang Zhao
- Medical College of Soochow University, Suzhou 215123, China
| | - Jie Hong
- Medical College of Soochow University, Suzhou 215123, China
| | - Shen Cheng
- Medical College of Soochow University, Suzhou 215123, China
| | - Xiaohong Yu
- Medical College of Soochow University, Suzhou 215123, China
| | - Dong Liu
- Medical College of Soochow University, Suzhou 215123, China
| | - Bingqing Xu
- Medical College of Soochow University, Suzhou 215123, China
| | - Renping Hu
- Medical College of Soochow University, Suzhou 215123, China
| | - Qingqing Sun
- Medical College of Soochow University, Suzhou 215123, China
| | - Jie Cheng
- Medical College of Soochow University, Suzhou 215123, China
| | - Zhe Cheng
- Medical College of Soochow University, Suzhou 215123, China
| | - Suxin Gui
- Medical College of Soochow University, Suzhou 215123, China
| | - Fashui Hong
- Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
17
|
Crespo J, Sun H, Welling TH, Tian Z, Zou W. T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr Opin Immunol 2013; 25:214-21. [PMID: 23298609 DOI: 10.1016/j.coi.2012.12.003] [Citation(s) in RCA: 573] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 12/11/2012] [Accepted: 12/13/2012] [Indexed: 12/24/2022]
Abstract
Human tumors progress despite the presence of tumor associated antigen (TAA)-specific T cells. Many different molecular and cellular mechanisms contribute to the failure of T cells to eradicate the tumor. These include immune suppressive networks that impair ongoing T cell function and enable tumor escape. Recent studies have started to reveal the nature of effector T cells in the tumor microenvironment. In this article we discuss T cell anergy, exhaustion, senescence, and stemness, and review the phenotype of dysfunctional T cell subsets and the underlying molecular mechanisms in the tumor microenvironments. We suggest that targeting T cell dysfunctional mechanisms and introducing/promoting T cell stemness are important approaches to treat patients with cancer.
Collapse
Affiliation(s)
- Joel Crespo
- Department of Surgery, Ann Arbor, MI, United States
| | | | | | | | | |
Collapse
|
18
|
Tumor-induced CD8+ T-cell dysfunction in lung cancer patients. Clin Dev Immunol 2012; 2012:741741. [PMID: 23118782 PMCID: PMC3483679 DOI: 10.1155/2012/741741] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/28/2012] [Accepted: 09/04/2012] [Indexed: 12/21/2022]
Abstract
Lung cancer is the leading cause of cancer deaths worldwide and one of the most common types of cancers. The limited success of chemotherapy and radiotherapy regimes have highlighted the need to develop new therapies like antitumor immunotherapy. CD8+ T-cells represent a major arm of the cell-mediated anti-tumor response and a promising target for developing T-cell-based immunotherapies against lung cancer. Lung tumors, however, have been considered to possess poor immunogenicity; even so, lung tumor-specific CD8+ T-cell clones can be established that possess cytotoxicity against autologous tumor cells. This paper will focus on the alterations induced in CD8+ T-cells by lung cancer. Although memory CD8+ T-cells infiltrate lung tumors, in both tumor-infiltrating lymphocytes (TILs) and malignant pleural effusions, these cells are dysfunctional and the effector subset is reduced. We propose that chronic presence of lung tumors induces dysfunctions in CD8+ T-cells and sensitizes them to activation-induced cell death, which may be associated with the poor clinical responses observed in immunotherapeutic trials. Getting a deeper knowledge of the evasion mechanisms lung cancer induce in CD8+ T-cells should lead to further understanding of lung cancer biology, overcome tumor evasion mechanisms, and design improved immunotherapeutic treatments for lung cancer.
Collapse
|
19
|
Prado-Garcia H, Romero-Garcia S, Morales-Fuentes J, Aguilar-Cazares D, Lopez-Gonzalez JS. Activation-induced cell death of memory CD8+ T cells from pleural effusion of lung cancer patients is mediated by the type II Fas-induced apoptotic pathway. Cancer Immunol Immunother 2012; 61:1065-80. [PMID: 22159518 PMCID: PMC11028981 DOI: 10.1007/s00262-011-1165-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 11/15/2011] [Indexed: 12/21/2022]
Abstract
Lung cancer is the second most common form of cancer and the leading cause of cancer death worldwide. Pleural effusions, containing high numbers of mononuclear and tumor cells, are frequent in patients with advanced stages of lung cancer. We reported that in pleural effusions from primary lung cancer, the CD8+ T cell subpopulation, and particularly the terminally differentiated subset, is reduced compared to that of non-malignant effusions. We analyzed the participation of activation-induced cell death (AICD) and extrinsic pathways (type I or II) as mechanisms for the decrease in pleural effusion CD8+ T cell subpopulation. Pleural effusion or peripheral blood CD4+ and CD8+ T cells, from lung cancer patients, were stimulated with anti-CD3 antibody and analyzed for (a) apoptosis by annexin-V-binding and TUNEL assay, (b) transcript levels of Fas ligand (FasL) and TRAIL by real-time RT-PCR, (c) expression of FasL and TRAIL, measured as integrated mean fluorescence intensities (iMFI) by flow cytometry, (d) expression of Bcl-2 and BIM molecules, measured as MFI, and (e) apoptosis inhibition using caspase-8 and -9 inhibitors. Pleural effusion CD8+ T cells, but not CD4+ T cells, from cancer patients underwent AICD. Blocking FasL/Fas pathway protected from AICD. Upregulation of FasL and TRAIL expressions was found in pleural effusion CD8+ T cells, which also showed a subset of Bcl-2 low cells. In memory CD8+ T cells, AICD depended on both extrinsic and intrinsic apoptotic pathways. Hence, in the pleural space of lung cancer patients, AICD might compromise the antitumor function of CD8+ T cells.
Collapse
Affiliation(s)
- Heriberto Prado-Garcia
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico.
| | | | | | | | | |
Collapse
|
20
|
Lehman HK, Simpson-Abelson MR, Conway TF, Kelleher RJ, Bernstein JM, Bankert RB. Memory T cells in the chronic inflammatory microenvironment of nasal polyposis are hyporesponsive to signaling through the T cell receptor. J Assoc Res Otolaryngol 2012; 13:423-35. [PMID: 22310933 PMCID: PMC3346897 DOI: 10.1007/s10162-012-0313-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 01/10/2012] [Indexed: 12/17/2022] Open
Abstract
A majority of T cells from chronic inflammatory tissues derived from patients with nasal polyposis were found to express an effector memory phenotype. We report here that these memory T cells failed to activate NF-κB in response to TCR stimulation but responded normally when the proximal TCR signaling molecules were bypassed with PMA and ionomycin. The dysfunction of these cells was associated with a decrease in the phosphorylation of several TCR proximal signaling molecules including ZAP70, Lck and SLP-76. In addition to the disruption in the TCR signaling pathway, the nasal polyp-associated T cells were shown to have a defect in their ability to translocate LAMP-1 to the cell surface. The results presented here establish that the phenotype and anergy of the T cells in the nasal polyp are similar to those which is seen in memory T cells derived from human tumors and other sites of chronic inflammation.
Collapse
Affiliation(s)
- Heather K. Lehman
- Department of Pediatrics, University at Buffalo School of Medicine and Biomedical Sciences, 239 Bryant St., 2nd Floor, Buffalo, NY 14222 USA
| | - Michelle R. Simpson-Abelson
- Department of Microbiology and Immunology, University at Buffalo School at Medicine and Biomedical Sciences, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
- Department of Immunology, University of Pittsburgh, S708 BST South, Pittsburgh, PA 15261 USA
| | - Thomas F. Conway
- Department of Microbiology and Immunology, University at Buffalo School at Medicine and Biomedical Sciences, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Raymond J. Kelleher
- Department of Microbiology and Immunology, University at Buffalo School at Medicine and Biomedical Sciences, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Joel M. Bernstein
- Department of Otolaryngology, University at Buffalo School of Medicine and Biomedical Sciences, 3435 Main Street, Buffalo, NY 14214 USA
| | - Richard B. Bankert
- Department of Microbiology and Immunology, University at Buffalo School at Medicine and Biomedical Sciences, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| |
Collapse
|
21
|
Bankert RB, Balu-Iyer SV, Odunsi K, Shultz LD, Kelleher RJ, Barnas JL, Simpson-Abelson M, Parsons R, Yokota SJ. Humanized mouse model of ovarian cancer recapitulates patient solid tumor progression, ascites formation, and metastasis. PLoS One 2011; 6:e24420. [PMID: 21935406 PMCID: PMC3174163 DOI: 10.1371/journal.pone.0024420] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/08/2011] [Indexed: 01/13/2023] Open
Abstract
Ovarian cancer is the most common cause of death from gynecological cancer. Understanding the biology of this disease, particularly how tumor-associated lymphocytes and fibroblasts contribute to the progression and metastasis of the tumor, has been impeded by the lack of a suitable tumor xenograft model. We report a simple and reproducible system in which the tumor and tumor stroma are successfully engrafted into NOD-scid IL2Rγnull (NSG) mice. This is achieved by injecting tumor cell aggregates derived from fresh ovarian tumor biopsy tissues (including tumor cells, and tumor-associated lymphocytes and fibroblasts) i.p. into NSG mice. Tumor progression in these mice closely parallels many of the events that are observed in ovarian cancer patients. Tumors establish in the omentum, ovaries, liver, spleen, uterus, and pancreas. Tumor growth is initially very slow and progressive within the peritoneal cavity with an ultimate development of tumor ascites, spontaneous metastasis to the lung, increasing serum and ascites levels of CA125, and the retention of tumor-associated human fibroblasts and lymphocytes that remain functional and responsive to cytokines for prolonged periods. With this model one will be able to determine how fibroblasts and lymphocytes within the tumor microenvironment may contribute to tumor growth and metastasis, and will make it possible to evaluate the efficacy of therapies that are designed to target these cells in the tumor stroma.
Collapse
Affiliation(s)
- Richard B Bankert
- Department of Microbiology and Immunology, The State University of New York, University at Buffalo, Buffalo, New York, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang SF, Fouquet S, Chapon M, Salmon H, Regnier F, Labroquère K, Badoual C, Damotte D, Validire P, Maubec E, Delongchamps NB, Cazes A, Gibault L, Garcette M, Dieu-Nosjean MC, Zerbib M, Avril MF, Prévost-Blondel A, Randriamampita C, Trautmann A, Bercovici N. Early T cell signalling is reversibly altered in PD-1+ T lymphocytes infiltrating human tumors. PLoS One 2011; 6:e17621. [PMID: 21408177 PMCID: PMC3049782 DOI: 10.1371/journal.pone.0017621] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 02/02/2011] [Indexed: 12/18/2022] Open
Abstract
To improve cancer immunotherapy, a better understanding of the weak efficiency of tumor-infiltrating T lymphocytes (TIL) is necessary. We have analyzed the functional state of human TIL immediately after resection of three types of tumors (NSCLC, melanoma and RCC). Several signalling pathways (calcium, phosphorylation of ERK and Akt) and cytokine secretion are affected to different extents in TIL, and show a partial spontaneous recovery within a few hours in culture. The global result is an anergy that is quite distinct from clonal anergy induced in vitro, and closer to adaptive tolerance in mice. PD-1 (programmed death -1) is systematically expressed by TIL and may contribute to their anergy by its mere expression, and not only when it interacts with its ligands PD-L1 or PD-L2, which are not expressed by every tumor. Indeed, the TCR-induced calcium and ERK responses were reduced in peripheral blood T cells transfected with PD-1. Inhibition by sodium stibogluconate of the SHP-1 and SHP-2 phosphatases that associate with several inhibitory receptors including PD-1, relieves part of the anergy apparent in TIL or in PD-1-transfected T cells. This work highlights some of the molecular modifications contributing to functional defects of human TIL.
Collapse
Affiliation(s)
- Shu-Fang Wang
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Stéphane Fouquet
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Maxime Chapon
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Hélène Salmon
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Fabienne Regnier
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Karine Labroquère
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Cécile Badoual
- Inserm U970, Univ Paris Descartes, PARCC, Paris, France
- Service d'Anatomie-Pathologique, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Diane Damotte
- Laboratoire Microenvironnement immunitaire et tumeurs, INSERM U872, Centre de Recherche des Cordeliers, Paris, France
- Univ Pierre et Marie Curie, UMR S872, Paris, France
- Univ Paris Descartes, UMR S872, Paris, France
- Service d'Anatomie-Pathologie, Hôpital Hôtel Dieu, AP-HP, Paris, France
| | - Pierre Validire
- Service d'Anatomie-Pathologie, Institut Mutualiste Montsouris, Paris, France
| | - Eve Maubec
- APHP, UnivParis Diderot, Service de Dermatologie, Hôpital Bichat, Paris, France
| | | | - Aurélie Cazes
- Service d'Anatomie-Pathologique, Hôpital Européen Georges Pompidou, APHP, Paris, France
- Inserm U833, Collège de France, Université Paris Descartes, Paris, France
| | - Laure Gibault
- Service d'anatomie et cytologie pathologiques, Groupe Hospitalier Cochin-Saint Vincent de Paul, Univ Paris Descartes, Paris, France
| | - Marylène Garcette
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- Laboratoire Microenvironnement immunitaire et tumeurs, INSERM U872, Centre de Recherche des Cordeliers, Paris, France
- Univ Pierre et Marie Curie, UMR S872, Paris, France
- Univ Paris Descartes, UMR S872, Paris, France
| | - Marc Zerbib
- APHP, Hôpital Cochin, service d'Urologie, Paris, France
| | - Marie-Françoise Avril
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
- APHP, Hôpital Cochin, Service de Dermatologie, Paris, France
| | - Armelle Prévost-Blondel
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Clotilde Randriamampita
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Alain Trautmann
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
- * E-mail: (AT); (NB)
| | - Nadège Bercovici
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
- * E-mail: (AT); (NB)
| |
Collapse
|
23
|
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: 12] [Impact Index Per Article: 0.8] [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
|
24
|
Barnas JL, Simpson-Abelson MR, Yokota SJ, Kelleher RJ, Bankert RB. T cells and stromal fibroblasts in human tumor microenvironments represent potential therapeutic targets. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2010; 3:29-47. [PMID: 21209773 PMCID: PMC2990491 DOI: 10.1007/s12307-010-0044-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 03/02/2010] [Indexed: 12/16/2022]
Abstract
The immune system of cancer patients recognizes tumor-associated antigens expressed on solid tumors and these antigens are able to induce tumor-specific humoral and cellular immune responses. Diverse immunotherapeutic strategies have been used in an attempt to enhance both antibody and T cell responses to tumors. While several tumor vaccination strategies significantly increase the number of tumor-specific lymphocytes in the blood of cancer patients, most vaccinated patients ultimately experience tumor progression. CD4+ and CD8+ T cells with an effector memory phenotype infiltrate human tumor microenvironments, but most are hyporesponsive to stimulation via the T cell receptor (TCR) and CD28 under conditions that activate memory T cells derived from the peripheral blood of the cancer patients or normal donors. Attempts to identify cells and molecules responsible for the TCR signaling arrest of tumor-infiltrating T cells have focused largely upon the immunosuppressive effects of tumor cells, tolerogenic dendritic cells and regulatory T cells. Here we review potential mechanisms by which human T cell function is arrested in the tumor microenvironment with a focus on the immunomodulatory effects of stromal fibroblasts. Determining in vivo which cells and molecules are responsible for the TCR arrest in human tumor-infiltrating T cells will be necessary to formulate and test strategies to prevent or reverse the signaling arrest of the human T cells in situ for a more effective design of tumor vaccines. These questions are now addressable using novel human xenograft models of tumor microenvironments.
Collapse
Affiliation(s)
- Jennifer L. Barnas
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Michelle R. Simpson-Abelson
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Sandra J. Yokota
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Raymond J. Kelleher
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Richard B. Bankert
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| |
Collapse
|
25
|
di Bari MG, Lutsiak MEC, Takai S, Mostböck S, Farsaci B, Tolouei Semnani R, Wakefield LM, Schlom J, Sabzevari H. TGF-beta modulates the functionality of tumor-infiltrating CD8+ T cells through effects on TCR signaling and Spred1 expression. Cancer Immunol Immunother 2009; 58:1809-18. [PMID: 19319531 PMCID: PMC3499107 DOI: 10.1007/s00262-009-0692-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 03/04/2009] [Indexed: 01/17/2023]
Abstract
This study demonstrates that CD8+ T cells in the tumor microenvironment display reduced functionality and hyporesponsiveness. TGF-beta contributed markedly to the tumor-infiltrating CD8+ T cells' (TILs) reduced functionality, which could be reversed using a small molecule TGF-beta inhibitor. Upon T-cell receptor (TCR) activation, the activation of ITK and ERK kinases were reduced in CD8+ TILs, as compared to splenic CD8+ T cells: TGF-beta inhibitor could reverse this phenomenon. This study demonstrates for the first time the association of the Spred-1 gene, an inhibitor of the Ras/MAPK pathway, with CD8+ TILs and TGF-beta activity. Spred-1 was upregulated in CD8+ TILs and TGF-beta enhanced the expression of Spred-1 in effector/memory CD8+ T cells and not in rested/memory CD8+ T cells. Based on these findings, this study supports the hypothesis that TGF-beta mediates an inhibitory mechanism on CD8+ TILs involving TCR-signaling blockade and the upregulation of Spred-1, thus implicating Spred-1 as a potential new target for future anti-tumor immune studies.
Collapse
Affiliation(s)
- Maria Giovanna di Bari
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD 20892 USA
| | - M. E. Christine Lutsiak
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD 20892 USA
| | - Shinji Takai
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD 20892 USA
| | - Sven Mostböck
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD 20892 USA
| | - Benedetto Farsaci
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD 20892 USA
| | - Roshanak Tolouei Semnani
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Lalage M. Wakefield
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD 20892 USA
| | - Helen Sabzevari
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, MSC 1750, Bethesda, MD 20892 USA
| |
Collapse
|
26
|
Simpson-Abelson MR, Purohit VS, Pang WM, Iyer V, Odunsi K, Demmy TL, Yokota SJ, Loyall JL, Kelleher RJ, Balu-Iyer S, Bankert RB. IL-12 delivered intratumorally by multilamellar liposomes reactivates memory T cells in human tumor microenvironments. Clin Immunol 2009; 132:71-82. [PMID: 19395317 PMCID: PMC2693480 DOI: 10.1016/j.clim.2009.03.516] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 02/16/2009] [Accepted: 03/17/2009] [Indexed: 01/07/2023]
Abstract
Using a novel loading technique, IL-12 is reported here to be efficiently encapsulated within large multilamellar liposomes. The preclinical efficacy of the cytokine loaded liposomes to deliver IL-12 into human tumors and to reactive tumor-associated T cells in situ is tested using a human tumor xenograft model. IL-12 is released in vivo from these liposomes in a biologically active form when injected into tumor xenografts that are established by the subcutaneous implantation of non-disrupted pieces of human lung, breast or ovarian tumors into immunodeficient mice. The histological architecture of the original tumor tissue, including tumor-associated leukocytes, tumor cells and stromal cells is preserved anatomically and the cells remain functionally responsive to cytokines in these xenografts. The local and sustained release of IL-12 into the tumor microenvironment reactivates tumor-associated quiescent effector memory T cells to proliferate, produce and release IFN-gamma resulting in the killing of tumor cells in situ. Very little IL-12 is detected in the serum of mice for up to 5 days after an intratumoral injection of the IL-12 liposomes. We conclude that IL-12 loaded large multilamellar liposomes provide a safe method for the local and sustained delivery of IL-12 to tumors and a therapeutically effective way of reactivating existing tumor-associated T cells in human solid tumor microenvironments. The potential of this local in situ T cell re-stimulation to induce a systemic anti-tumor immunity is discussed.
Collapse
Affiliation(s)
- Michelle R. Simpson-Abelson
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Vivek S. Purohit
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Wing Man Pang
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Vandana Iyer
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Todd L Demmy
- Department of Thoracic Surgery, Roswell Park Cancer Institute, Buffalo, New York
| | - Sandra J. Yokota
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Jenni L. Loyall
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Raymond J. Kelleher
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Sathy Balu-Iyer
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Richard B. Bankert
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| |
Collapse
|
27
|
Airoldi I, Di Carlo E, Cocco C, Caci E, Cilli M, Sorrentino C, Sozzi G, Ferrini S, Rosini S, Bertolini G, Truini M, Grossi F, Galietta LJV, Ribatti D, Pistoia V. IL-12 can target human lung adenocarcinoma cells and normal bronchial epithelial cells surrounding tumor lesions. PLoS One 2009; 4:e6119. [PMID: 19582164 PMCID: PMC2702099 DOI: 10.1371/journal.pone.0006119] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 05/21/2009] [Indexed: 01/09/2023] Open
Abstract
Background Non small cell lung cancer (NSCLC) is a leading cause of cancer death. We have shown previously that IL-12rb2 KO mice develop spontaneously lung adenocarcinomas or bronchioalveolar carcinomas. Aim of the study was to investigate i) IL-12Rβ2 expression in human primary lung adenocarcinomas and in their counterparts, i.e. normal bronchial epithelial cells (NBEC), ii) the direct anti-tumor activity of IL-12 on lung adenocarcinoma cells in vitro and vivo, and the mechanisms involved, and iii) IL-12 activity on NBEC. Methodology/Principal Findings Stage I lung adenocarcinomas showed significantly (P = 0.012) higher frequency of IL-12Rβ2 expressing samples than stage II/III tumors. IL-12 treatment of IL-12R+ neoplastic cells isolated from primary adenocarcinoma (n = 6) inhibited angiogenesis in vitro through down-regulation of different pro-angiogenic genes (e.g. IL-6, VEGF-C, VEGF-D, and laminin-5), as assessed by chorioallantoic membrane (CAM) assay and PCR array. In order to perform in vivo studies, the Calu6 NSCLC cell line was transfected with the IL-12RB2 containing plasmid (Calu6/β2). Similar to that observed in primary tumors, IL-12 treatment of Calu6/β2+ cells inhibited angiogenesis in vitro. Tumors formed by Calu6/β2 cells in SCID/NOD mice, inoculated subcutaneously or orthotopically, were significantly smaller following IL-12 vs PBS treatment due to inhibition of angiogenesis, and of IL-6 and VEGF-C production. Explanted tumors were studied by histology, immuno-histochemistry and PCR array. NBEC cells were isolated and cultured from lung specimens of non neoplastic origin. NBEC expressed IL-12R and released constitutively tumor promoting cytokines (e.g. IL-6 and CCL2). Treatment of NBEC with IL-12 down-regulated production of these cytokines. Conclusions This study demonstrates that IL-12 inhibits directly the growth of human lung adenocarcinoma and targets the adjacent NBEC. These novel anti-tumor activities of IL-12 add to the well known immune-modulatory properties of the cytokine and may provide a rational basis for the development of a clinical trial.
Collapse
Affiliation(s)
- Irma Airoldi
- AIRC Tumor Immunology Unit, Department of Experimental and Laboratory Medicine, G Gaslini Institute, Genova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Memory T cells exhibit low activation thresholds and rapid effector responses following antigen stimulation, contrasting naive T cells with high activation thresholds and no effector responses. Signaling mechanisms for the distinct properties of naive and memory T cells remain poorly understood. Here, I will discuss new results on signal transduction in naive and memory T cells that suggest proximal control of activation threshold and a distinct biochemical pathway to rapid recall. The signaling and transcriptional pathways controlling immediate effector function in memory T cells closely resemble pathways for rapid effector cytokine production in innate immune cells, suggesting memory T cells use innate pathways for efficacious responses.
Collapse
Affiliation(s)
- Donna L Farber
- Department of Surgery, University of Maryland School of Medicine, MSTF Building, Room 400, 685 W. Baltimore Street, Baltimore, MD 21201, United States.
| |
Collapse
|
29
|
Derniame S, Vignaud JM, Faure GC, Béné MC. Alteration of the immunological synapse in lung cancer: a microenvironmental approach. Clin Exp Immunol 2008; 154:48-55. [PMID: 18761663 DOI: 10.1111/j.1365-2249.2008.03727.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study was designed to investigate the immunological properties of stroma reaction T cells and tumoral cells by comparison with non-tumoral lung tissue and local lymph nodes in order to explore interactions between tumour cells and the immune system. Immunodetection of major histocompatibility complex (MHC) molecules, CD3/T cell receptor (TCR) complex and T cell subsets markers was carried out in situ on frozen sections, and the semi-quantitative expression of CD3, CD4 and CD8 was examined in flow cytometry on lymphocytes of nodal, tumoral and healthy lung tissue from 62 patients with non-small cell lung cancer. This study showed alterations on lymphocytes and tumour cells in lung cancer, consistent with an impairment of T cell activation. CD3, TCR alpha beta and accessory molecules expression is down-modulated on peri- or intra-tumoral lymphocytes. MHC class I and class II molecules are down-modulated significantly on tumour cells. Other differences were noted, such as the reversed CD4/CD8 ratio of tumour infiltrating cells, compared to healthy lung tissues, consistent with the development of cytotoxic anti-tumoral responses. This study reports on the presence of a strong in vivo immunomodulating effect of tumour cells in human non-small cell lung cancer, likely to impair proper formation of the immunological synapse.
Collapse
Affiliation(s)
- S Derniame
- Laboratoire d'Immunologie, Faculté de Médecine et CHU de Nancy, Vandoeuvre les Nancy, Nancy, France.
| | | | | | | |
Collapse
|
30
|
Kroening PR, Barnes TW, Pease L, Limper A, Kita H, Vassallo R. Cigarette smoke-induced oxidative stress suppresses generation of dendritic cell IL-12 and IL-23 through ERK-dependent pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:1536-47. [PMID: 18606709 PMCID: PMC2819390 DOI: 10.4049/jimmunol.181.2.1536] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
IL-12p70, a heterodimer composed of p35 and p40 subunits, is a key polarizing cytokine produced by maturing dendritic cells (DCs). We report that cigarette smoke extract (CSE), an extract of soluble cigarette smoke components, suppresses both p35 and p40 production by LPS or CD40L-matured DCs. Suppression of IL-12p70 production from maturing DCs was not observed in the presence of nicotine concentrations achievable in CSE or in the circulation of smokers. The suppressed IL-12p70 protein production by CSE-conditioned DCs was restored by pretreatment of DCs or CSE with the antioxidants N-acetylcysteine and catalase. Inhibition of DC IL-12p70 by CSE required activation of ERK-dependent pathways, since inhibition of ERK abrogated the suppressive effect of CSE on IL-12 secretion. Oxidative stress and sustained ERK phosphorylation by CSE enhanced nuclear levels of the p40 transcriptional repressor c-fos in both immature and maturing DCs. Suppression of the p40 subunit by CSE also resulted in diminished production of IL-23 protein by maturing DCs. Using a murine model of chronic cigarette smoke exposure, we observed that systemic and lung DCs from mice "smokers" produced significantly less IL-12p70 and p40 protein upon maturation. This inhibitory effect was selective, since production of TNF-alpha during DC maturation was enhanced in the smokers. These data imply that oxidative stress generated by cigarette smoke exposure suppresses the generation of key cytokines by maturing DCs through the activation of ERK-dependent pathways. Some of the cigarette smoke-induced inhibitory effects on DC function may be mitigated by antioxidants.
Collapse
Affiliation(s)
- Paula R Kroening
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care, Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA
| | | | - Larry Pease
- Department of Immunology, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA
| | - Andrew Limper
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care, Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA
| | - Hirohito Kita
- Department of Immunology, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA
- Division of Allergic diseases, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA
| | - Robert Vassallo
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care, Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA
| |
Collapse
|
31
|
Simpson-Abelson MR, Sonnenberg GF, Takita H, Yokota SJ, Conway TF, Kelleher RJ, Shultz LD, Barcos M, Bankert RB. Long-term engraftment and expansion of tumor-derived memory T cells following the implantation of non-disrupted pieces of human lung tumor into NOD-scid IL2Rgamma(null) mice. THE JOURNAL OF IMMUNOLOGY 2008; 180:7009-18. [PMID: 18453623 DOI: 10.4049/jimmunol.180.10.7009] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Non-disrupted pieces of primary human lung tumor implanted into NOD-scid IL2Rgamma(null) mice consistently result in successful xenografts in which tissue architecture, including tumor-associated leukocytes, stromal fibroblasts, and tumor cells are preserved for prolonged periods with limited host-vs-graft interference. Human CD45(+) tumor-associated leukocytes within the xenograft are predominantly CD3(+) T cells with fewer CD138(+) plasma cells. The effector memory T cells that had been shown to be quiescent in human lung tumor microenvironments can be activated in situ as determined by the production of human IFN-gamma in response to exogenous IL-12. Plasma cells remain functional as evidenced by production of human Ig. Significant levels of human IFN-gamma and Ig were detected in sera from xenograft-bearing mice for up to 9 wk postengraftment. Tumor-associated T cells were found to migrate from the microenvironment of the xenograft to the lung, liver, and primarily the spleen. At 8 wk postengraftment, a significant portion of cells isolated from the mouse spleens were found to be human CD45(+) cells. The majority of CD45(+) cells were CD3(+) and expressed a phenotype consistent with an effector memory T cell, consisting of CD4(+) or CD8(+) T cells that were CD45RO(+), CD44(+), CD62L(-), and CD25(-). Following adoptive transfer into non-tumor bearing NOD-scid IL2Rgamma(null) mice, these human T cells were found to expand in the spleen, produce IFN-gamma, and maintain an effector memory phenotype. We conclude that the NOD-scid IL2Rgamma(null) tumor xenograft model provides an opportunity to study tumor and tumor-stromal cell interactions in situ for prolonged periods.
Collapse
Affiliation(s)
- Michelle R Simpson-Abelson
- State University of New York, Department of Microbiology and Immunology and the Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, Buffalo, NY 14214, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Simpson-Abelson M, Bankert RB. Targeting the TCR signaling checkpoint: a therapeutic strategy to reactivate memory T cells in the tumor microenvironment. Expert Opin Ther Targets 2008; 12:477-90. [DOI: 10.1517/14728222.12.4.477] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
33
|
Abstract
Substantial evidence shows that inflammation promotes oncogenesis and, occasionally, participates in cancer rejection. This paradox can be accounted for by a dynamic switch from chronic smouldering inflammation promoting cancer-cell survival to florid, tissue-disruptive inflammatory reactions that trigger cancer-cell destruction. Clinical and experimental observations suggest that the mechanism of this switch recapitulates the events associated with pathogen infection, which stimulate immune cells to recognise danger signals and activate immune effector functions. Generally, cancers do not have danger signals and, therefore, they cannot elicit strong immune reactions. Synthetic molecules have been developed that mimic pathogen invasion at the tumour site. These compounds activate dendritic cells to produce proinflammatory cytokines, which in turn trigger cytotoxic mechanisms leading to cancer death. Simultaneously, dendritic cells capture antigen shed by dying cancer cells, undergo activation, and stimulate antigen-specific T and B cells. This process results in massive amplification of the antineoplastic inflammatory process. Thus, although anti-inflammatory drugs can prevent onset of some malignant diseases, induction of T cells specific for tumour antigen by active immunisation, combined with powerful activation signals within the cancer microenvironment, might yield the best strategy for treatment of established cancers.
Collapse
Affiliation(s)
- Alberto Mantovani
- Istituto Clinico Humanitas and Institute of Pathology, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
34
|
Prado-Garcia H, Aguilar-Cazares D, Meneses-Flores M, Morales-Fuentes J, Lopez-Gonzalez JS. Lung carcinomas do not induce T-cell apoptosis via the Fas/Fas ligand pathway but down-regulate CD3 epsilon expression. Cancer Immunol Immunother 2008; 57:325-36. [PMID: 17668204 PMCID: PMC11030893 DOI: 10.1007/s00262-007-0372-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 07/11/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND Non-small cell lung carcinoma (NSCLC) patients have impaired cellular immune responses. It has been hypothesized that tumor cells expressing Fas Ligand (FasL) induce in T lymphocytes: (a) apoptosis (tumor counterattack) and (b) down-regulation of CD3zeta expression. However, the hypothesis of tumor counterattack is still controversial. METHODS We analyzed FasL expression on NSCLC cell lines and on tumor cells from lung adenocarcinoma patients by flow cytometry and immunocytochemistry. FasL mRNA expression was detected in NSCLC cell lines using RT-PCR, and functional FasL was evaluated on Fas-expressing Jurkat T-cells by annexin-V-FITC staining and by SubG(1) peak detection. Also, the proapoptotic effect of microvesicles released from NSCLC cell lines in Jurkat T-cells was studied. Alterations in the expression levels of CD3zeta, CD3epsilon, and CD28 [measured as mean fluorescence intensity (MFI)] were determined in Jurkat T-cells after co-culture with NSCLC cell lines or tumor-derived microvesicles. Furthermore, the expression levels of CD3zeta and CD3epsilon in CD4+T and CD8+T lymphocytes from lung adenocarcinoma patients was studied. RESULTS Our results indicate that NSCLC cells neither FasL expressed nor induced apoptosis in Jurkat T-cells. Tumor-derived microvesicles did not induce apoptosis in Jurkat T-cells. In contrast, NSCLC cell lines down-regulated CD3epsilon but not CD3zeta chain expression in Jurkat T-cells; this effect was induced by soluble factors but not by microvesicles. In lung adenocarcinoma patients, significant decreases of MFI values for CD3epsilon, but not CD3zeta, were found in CD4+T and CD8+T cells from pleural effusion compared to peripheral blood and in peripheral blood of patients compared to healthy donors. CONCLUSIONS Our data do not support the tumor counterattack hypothesis for NSCLC. Nonetheless, down-regulation of CD3epsilon in T-cells induced by NSCLC cells might lead to T-cell dysfunction.
Collapse
Affiliation(s)
- Heriberto Prado-Garcia
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Dolores Aguilar-Cazares
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Manuel Meneses-Flores
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Jorge Morales-Fuentes
- Servicio Clinico 3, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Jose Sullivan Lopez-Gonzalez
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| |
Collapse
|
35
|
Nazareth MR, Broderick L, Simpson-Abelson MR, Kelleher RJ, Yokota SJ, Bankert RB. Characterization of human lung tumor-associated fibroblasts and their ability to modulate the activation of tumor-associated T cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:5552-62. [PMID: 17442937 DOI: 10.4049/jimmunol.178.9.5552] [Citation(s) in RCA: 208] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The tumor microenvironment of human non-small cell lung cancer (NSCLC) is composed largely of stromal cells, including fibroblasts, yet these cells have been the focus of few studies. In this study, we established stromal cell cultures from primary NSCLC through isolation of adherent cells. Characterization of these cells by flow cytometry demonstrated a population which expressed a human fibroblast-specific 112-kDa surface molecule, Thy1, alpha-smooth muscle actin, and fibroblast activation protein, but failed to express CD45 and CD11b, a phenotype consistent with that of an activated myofibroblast. A subset of the tumor-associated fibroblasts (TAF) was found to express B7H1 (PD-L1) and B7DC (PD-L2) constitutively, and this expression was up-regulated by IFN-gamma. Production of cytokines and chemokines, including IFN-gamma, monokine induced by IFN-gamma, IFN-gamma-inducible protein-10, RANTES, and TGF-beta1 was also demonstrated in these cells. Together, these characteristics provide multiple opportunities for the TAF to influence cellular interactions within the tumor microenvironment. To evaluate the ability of TAF to modulate tumor-associated T cell (TAT) activation, we conducted coculture experiments between autologous TAF and TAT. In five of eight tumors, TAF elicited a contact-dependent enhancement of TAT activation, even in the presence of a TGF-beta1-mediated suppressive effect. In the three other tumors, TAF had a net suppressive effect upon TAT activation, and, in one of these cases, blockade of B7H1 or B7DC was able to completely abrogate the TAF-mediated suppression. We conclude that TAF in human NSCLC are functionally and phenotypically heterogeneous and provide multiple complex regulatory signals that have the potential to enhance or suppress TAT function in the tumor microenvironment.
Collapse
MESH Headings
- Antigens, CD/analysis
- Antigens, CD/metabolism
- B7-1 Antigen/analysis
- B7-1 Antigen/metabolism
- B7-H1 Antigen
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Chemokines/metabolism
- Coculture Techniques
- Cytokines/metabolism
- Fibroblasts/immunology
- Fibroblasts/pathology
- Humans
- Interferon-gamma/metabolism
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lymphocyte Activation
- Programmed Cell Death 1 Ligand 2 Protein
- Receptors, Antigen, T-Cell/agonists
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- Transforming Growth Factor beta/analysis
- Transforming Growth Factor beta/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Michael R Nazareth
- Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, State University of New York, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
The cornerstone of the concept of immunosurveillance in cancer should be the experimental demonstration of immune responses able to alter the course of in vivo spontaneous tumor progression. Elegant genetic manipulation of the mouse immune system has proved this tenet. In parallel, progress in understanding human T cell mediated immunity has allowed to document the existence in cancer patients of naturally acquired T cell responses to molecularly defined tumor antigens. Various attributes of cutaneous melanoma tumors, notably their adaptability to in vitro tissue culture conditions, have contributed to convert this tumor in the prototype for studies of human antitumor immune responses. As a consequence, the first human cytolytic T lymphocyte (CTL)-defined tumor antigen and numerous others have been identified using lymphocyte material from patients bearing this tumor, detailed analyses of specific T cell responses have been reported and a relatively large number of clinical trials of vaccination have been performed in the last 15 years. Thus, the "melanoma model" continues to provide valuable insights to guide the development of clinically effective cancer therapies based on the recruitment of the immune system. This chapter reviews recent knowledge on human CD8 and CD4 T cell responses to melanoma antigens.
Collapse
Affiliation(s)
- Pedro Romero
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, University Hospital (CHUV), Lausanne, Switzerland
| | | | | |
Collapse
|
37
|
Broderick L, Bankert RB. Memory T cells in human tumor and chronic inflammatory microenvironments: sleeping beauties re-awakened by a cytokine kiss. Immunol Invest 2006; 35:419-36. [PMID: 16916760 DOI: 10.1080/08820130600755066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human tumors often progress and spread in spite of the presence of large numbers of CD4+ and CD8+ T cells with activated or memory cell phenotypes. The T cells in the microenvironment of human lung tumors fail to be activated in response to stimulation via the T cell receptor and CD28 under conditions that fully activate T cells derived from the peripheral blood of the cancer patients. A combination of regulatory mechanisms which are also observed in a variety of different chronic inflammatory conditions may contribute to the T cell unresponsiveness, and to their inability to respond to and kill tumor cells. The non-responsiveness of memory T cells isolated from human lung tumors and non-malignant chronic inflammatory tissues can be reversed in vitro by a brief pulse with IL-12, and the local and sustained release of exogenous IL-12 into the microenvironment of human tumor xenografts in SCID mice re-activates the tumor-associated T cells in situ. In the later case, the T cells proliferate, secrete interferon-gamma and initiate a cascade of events that culminate in the eradication of tumor cells from the xenograft. In transplantable and spontaneously developing tumors of mice the injection of a single tumor nodule with IL-12 loaded biodegradable microspheres activates tumor-associated T cells to kill tumor cells in situ, and provokes a systemic anti-tumor response that results in the eradication of distant metastatic tumor nodules that are not treated with the cytokine. These mice exhibit a systemic tumor specific immunity as they resist a second challenge with the same (but not a different) tumor. These findings suggest that it will be possible to provoke a systemic anti-tumor immunity in cancer patients by the direct injection of IL-12 loaded biodegradable microspheres or liposomes to locally deliver very low but sustained doses of IL-12 into a single tumor site. This strategy which is based upon the ability of IL-12 to re-activate tumor-associated T cells is termed in situ tumor vaccination.
Collapse
Affiliation(s)
- Lori Broderick
- Department of Microbiology and Immunology, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | | |
Collapse
|
38
|
Broderick L, Bankert RB. Membrane-associated TGF-beta1 inhibits human memory T cell signaling in malignant and nonmalignant inflammatory microenvironments. THE JOURNAL OF IMMUNOLOGY 2006; 177:3082-8. [PMID: 16920945 DOI: 10.4049/jimmunol.177.5.3082] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TGF-beta1 is present on cells derived from the microenvironment of human lung tumors and nonmalignant inflammatory tissues. We establish that this cell-associated cytokine mediates hyporesponsiveness of the memory T cells in these microenvironments in situ by blocking TCR signaling. T cells derived from these tissues failed to translocate NF-kappaB to the nucleus in response to CD3 + CD28 cross-linking. This nonresponsiveness was reversed by an anti-TGF-beta1-neutralizing Ab. Refractoriness of the memory T cells to TCR activation was also reversed by the removal of TGF-beta1 by briefly pulsing the cells in a low pH buffer. Addition of exogenous TGF-beta1 to eluted T cells re-established their nonresponsive state. Neither TGF-beta1, anti-TGF-beta1 Ab, nor low pH affected TCR signaling potential of peripheral blood T cells. We conclude that TGF-beta1 mediates a physiologically relevant regulatory mechanism, selective for memory T cells present in the tumor microenvironment and nonmalignant chronic inflammatory tissues.
Collapse
Affiliation(s)
- Lori Broderick
- Department of Microbiology and Immunology, State University of New York, Buffalo, NY 14214, USA
| | | |
Collapse
|