101
|
Abstract
The blockade of inhibitory receptors such as CTLA-4 (CD152) is being used as immune-checkpoint therapy, offering a powerful strategy to restore effective immune responses against tumors. To determine signal components that are induced under the control of CTLA-4 we analyzed activated murine CD8+ T cells by quantitative proteomics. Accurate mass spectrometry revealed that CTLA-4 engagement led to central changes in the phosphorylation of proteins involved in T-cell differentiation. Beside other targets, we discovered a CTLA-4-mediated induction of the translational inhibitor programmed cell death-4 (PDCD4) as a result of FoxO1 nuclear re-localization. PDCD4 further bound a distinct set of mRNAs including Glutaminase, which points out a critical role for CTLA-4 in CD8+ T-cell metabolism. Consequently, PDCD4-deficient cytotoxic T-lymphocytes (CTLs) expressed increased amounts of otherwise repressed effector molecules and ultimately led to superior control of tumor growth in vivo. These findings reveal a novel CTLA-4-mediated pathway to attenuate CTLs and indicate the importance of post-transcriptional mechanisms in the regulation of anti-tumor immune responses.
Collapse
|
102
|
Abstract
Skin is the largest organ of the body with a complex network of multitude of cell types that perform plastic and dynamic cellular communication to maintain several vital processes such as inflammation, immune response including induction of tolerance and disease prevention, wound healing, and angiogenesis. Of paramount importance are immunological functions of the skin that protect from harmful exposure coming from external and internal environments. Awareness of skin immunity can provide a better comprehension of inflammation, autoimmunity, cancer, graft-versus-host disease, vaccination, and immunotherapy approaches. This paper will update on what we currently know about immune sentinels contributing to skin immunity.
Collapse
Affiliation(s)
- Agata Matejuk
- Faculty of Health Science, Wroclaw Medical University, Wrocław, Poland. .,Faculty of Science and Technology, Karkonosze College, Jelenia Góra, Poland.
| |
Collapse
|
103
|
Wang J, Müller S, Lin R, Siffert M, Vuitton DA, Wen H, Gottstein B. Depletion of FoxP3 + Tregs improves control of larval Echinococcus multilocularis infection by promoting co-stimulation and Th1/17 immunity. IMMUNITY INFLAMMATION AND DISEASE 2017. [PMID: 28621034 PMCID: PMC5691311 DOI: 10.1002/iid3.181] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction The growth potential of the tumor‐like Echinococcus multilocularis metacestode (causing alveolar echinococcosis, AE) is directly linked to the nature/function of the periparasitic host immune‐mediated processes. Previous studies had shown that regulatory T cells (Tregs) become gradually up‐regulated in the course of both chronic human and murine AE. Thus we now tackled the role of FoxP3+ Tregs and FoxP3+‐Treg‐regulated immune response in contributing to the control of this helminthic infection. Methods The infection outcome in E. multilocularis‐infected DEREG mice was measured upon determining parasite load (wet weight of parasitic metacestode tissue). Flow cytometry and qRT‐PCR were used to assess Treg, Th17‐, Th1‐, Th2‐type immune responses and antigen presenting cell activation. Results We showed that E. multilocularis‐infected DEREG‐mice treated with DT (as compared to infected control DEREG‐mice without DT application) exhibited a significantly lower parasite load, associated with a persisting capacity of co‐stimulation, and an increased Th1/Th17‐polarization. Conclusions FoxP3+ Tregs appear as one of the key players in immune regulatory processes favoring (i) metacestode survival by inhibiting the maturation potential of co‐stimulatory activity and (ii) T cell exhaustion (suppressing Th1/Th17‐type immune responses). We showed as well that prospectively, targeting FoxP3+ Tregs could be an option to develop an immunotherapy against AE.
Collapse
Affiliation(s)
- Junhua Wang
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, Institute of Parasitology, University of Bern, Bern, Switzerland.,State Key Lab Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Stephan Müller
- FACSLab, c/o Institute of Pathology, University of Bern, Bern, Switzerland
| | - Renyong Lin
- State Key Lab Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Myriam Siffert
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, Central Animal Facilities, University of Bern, Bern, Switzerland
| | - Dominique A Vuitton
- WHO-Collaborating Centre on Prevention and Treatment of Human Echinococcosis and French National Reference Centre on Alveolar Echinococcosis, University of Franche-Comté and University Hospital, Besançon, France
| | - Hao Wen
- State Key Lab Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Bruno Gottstein
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, Institute of Parasitology, University of Bern, Bern, Switzerland
| |
Collapse
|
104
|
Heine A, Flores C, Gevensleben H, Diehl L, Heikenwalder M, Ringelhan M, Janssen KP, Nitsche U, Garbi N, Brossart P, Knolle PA, Kurts C, Höchst B. Targeting myeloid derived suppressor cells with all-trans retinoic acid is highly time-dependent in therapeutic tumor vaccination. Oncoimmunology 2017; 6:e1338995. [PMID: 28920004 DOI: 10.1080/2162402x.2017.1338995] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/28/2017] [Accepted: 06/01/2017] [Indexed: 12/30/2022] Open
Abstract
Tumor immune escape is a critical problem which frequently accounts for the failure of therapeutic tumor vaccines. Among the most potent suppressors of tumor immunity are myeloid derived suppressor cells (MDSCs). MDSCs can be targeted by all-trans-retinoic-acid (atRA), which reduced their numbers and increased response rates in several vaccination studies. However, not much is known about the optimal administration interval between atRA and the vaccine as well as about its mode of action. Here we demonstrate in 2 different murine tumor models that mice unresponsive to a therapeutic vaccine harbored higher MDSC numbers than did responders. Application of atRA overcame MDSC-mediated immunosuppression and restored tumor control. Importantly, atRA was protective only when administered 3 d after vaccination (delayed treatment), whereas simultaneous administration even decreased the anti-tumor immune response and reduced survival. When analyzing the underlying mechanisms, we found that delayed, but not simultaneous atRA treatment with vaccination abrogated the suppressive capacity in monocytic MDSCs and instead caused them to upregulate MHC-class-II. Consistently, MDSCs from patients with colorectal carcinoma also failed to upregulate HLA-DR after ex vivo treatment with TLR-ligation. Overall, we demonstrate that atRA can convert non-responders to responders to vaccination by suppressing MDSCs function and not only by reducing their number. Moreover, we identify a novel, strictly time-dependent mode of action of atRA to be considered during immunotherapeutic protocols in the future.
Collapse
Affiliation(s)
- Annkristin Heine
- Medical Clinic III for Oncology, Hematology and Rheumatology, University Hospital Bonn, Germany.,Institute of Experimental Immunology, University Bonn, Germany
| | - Chrystel Flores
- Medical Clinic III for Oncology, Hematology and Rheumatology, University Hospital Bonn, Germany.,Institute of Experimental Immunology, University Bonn, Germany
| | | | - Linda Diehl
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Germany.,Institute of Molecular Medicine, University Bonn, Germany
| | - Mathias Heikenwalder
- Institute of Virology, Technische Universität München.,Division of Chronic Inflammation and Cancer, German Cancer Research Center, DKFZ, Germany
| | - Marc Ringelhan
- Department for Internal Medicine 2, Klinikum rechts der Isar, Technische Universität München, Germany
| | | | - Ulrich Nitsche
- Department of Surgery, Technische Universität München, Germany
| | - Natalio Garbi
- Institute of Experimental Immunology, University Bonn, Germany
| | - Peter Brossart
- Medical Clinic III for Oncology, Hematology and Rheumatology, University Hospital Bonn, Germany
| | - Percy A Knolle
- Institute of Molecular Medicine, University Bonn, Germany.,Institute of Molecular Immunology and Experimental Oncology, Technische Universität München, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, University Bonn, Germany
| | - Bastian Höchst
- Institute of Molecular Medicine, University Bonn, Germany.,Institute of Molecular Immunology and Experimental Oncology, Technische Universität München, Germany
| |
Collapse
|
105
|
Ciurkiewicz M, Herder V, Khan MA, Uhde AK, Teich R, Floess S, Baumgärtner W, Huehn J, Beineke A. Cytotoxic CD8 + T cell ablation enhances the capacity of regulatory T cells to delay viral elimination in Theiler's murine encephalomyelitis. Brain Pathol 2017; 28:349-368. [PMID: 28452087 DOI: 10.1111/bpa.12518] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/18/2017] [Indexed: 12/28/2022] Open
Abstract
Theiler's murine encephalomyelitis (TME) of susceptible mouse strains is a commonly used infectious animal model for multiple sclerosis. The study aim was to test the hypothesis whether cytotoxic T cell responses account for the limited impact of regulatory T cells on antiviral immunity in TME virus-induced demyelinating disease (TMEV-IDD) resistant C57BL/6 mice. TME virus-infected C57BL/6 mice were treated with (i) interleukin-2/-anti-interleukin-2-antibody-complexes to expand regulatory T cells ("Treg-expansion"), (ii) anti-CD8-antibodies to deplete cytotoxic T cells ("CD8-depletion") or (iii) with a combination of Treg-expansion and CD8-depletion ("combined treatment") prior to infection. Results showed that "combined treatment", but neither sole "Treg-expansion" nor "CD8-depletion," leads to sustained hippocampal infection and virus spread to the spinal cord in C57BL/6 mice. Prolonged infection reduces myelin basic protein expression in the spinal cord together with increased accumulation of β-amyloid precursor protein in axons, characteristic of myelin loss and axonal damage, respectively. Chronic spinal cord infection upon "combined treatment" was also associated with increased T and B cell recruitment, accumulation of CD107b+ microglia/macrophages and enhanced mRNA expression of interleukin (IL)-1α, IL-10 and tumor necrosis factor α. In conclusion, data revealed that the suppressive capacity of Treg on viral elimination is efficiently boosted by CD8-depletion, which renders C57BL/6 mice susceptible to develop chronic neuroinfection and TMEV-IDD.
Collapse
Affiliation(s)
- Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Vanessa Herder
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Muhammad Akram Khan
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany.,Department of Pathobiology, Faculty of Veterinary & Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Ann-Kathrin Uhde
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - René Teich
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stephan Floess
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
106
|
Feng X, Zhang L, Acharya C, An G, Wen K, Qiu L, Munshi NC, Tai YT, Anderson KC. Targeting CD38 Suppresses Induction and Function of T Regulatory Cells to Mitigate Immunosuppression in Multiple Myeloma. Clin Cancer Res 2017; 23:4290-4300. [PMID: 28249894 DOI: 10.1158/1078-0432.ccr-16-3192] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/06/2017] [Accepted: 02/24/2017] [Indexed: 12/21/2022]
Abstract
Purpose: We study CD38 levels in immunosuppressive CD4+CD25highFoxp3+ regulatory T cells (Treg) and further define immunomodulating effects of a therapeutic CD38 mAb isatuximab/SAR650984 in multiple myeloma.Experimental Design: We evaluated percentages of CD38-expressing subsets in Tregs from normal donors and multiple myeloma patients. Peripheral blood mononuclear cells (PBMC) were then treated with isatuximab with or without lenalidomide or pomalidomide to identify their impact on the percentage and immunosuppressive activity of Tregs on CD4+CD25- T cells (Tcons). We investigated the mechanism of increased Tregs in multiple myeloma patients in ex vivo cocultures of multiple myeloma cells with PBMCs or Tcons.Results: CD38 expression is higher on Tregs than Tcons from multiple myeloma patients versus normal donors. CD38 levels and the percentages of CD38high Tregs are increased by lenalidomide and pomalidomide. Isatuximab preferentially decreases Treg and increases Tcon frequencies, which is enhanced by pomalidomide/lenalidomide. Isatuximab reduces Foxp3 and IL10 in Tregs and restores proliferation and function of Tcons. It augments multiple myeloma cell lysis by CD8+ T and natural killer cells. Coculture of multiple myeloma cells with Tcons significantly induces Tregs (iTregs), which express even higher CD38, CD25, and FoxP3 than natural Tregs. This is associated with elevated circulating CD38+ Tregs in multiple myeloma patients versus normal donors. Conversely, isatuximab decreases multiple myeloma cell- and bone marrow stromal cell-induced iTreg by inhibiting both cell-cell contact and TGFβ/IL10. Finally, CD38 levels correlate with differential inhibition by isatuximab of Tregs from multiple myeloma versus normal donors.Conclusions: Targeting CD38 by isatuximab can preferentially block immunosuppressive Tregs and thereby restore immune effector function against multiple myeloma. Clin Cancer Res; 23(15); 4290-300. ©2017 AACR.
Collapse
Affiliation(s)
- Xiaoyan Feng
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Li Zhang
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Chirag Acharya
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Gang An
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kenneth Wen
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Nikhil C Munshi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
107
|
Fisher SA, Aston WJ, Chee J, Khong A, Cleaver AL, Solin JN, Ma S, Lesterhuis WJ, Dick I, Holt RA, Creaney J, Boon L, Robinson B, Lake RA. Transient Treg depletion enhances therapeutic anti-cancer vaccination. Immun Inflamm Dis 2017; 5:16-28. [PMID: 28250921 PMCID: PMC5322183 DOI: 10.1002/iid3.136] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Regulatory T cells (Treg) play an important role in suppressing anti- immunity and their depletion has been linked to improved outcomes. To better understand the role of Treg in limiting the efficacy of anti-cancer immunity, we used a Diphtheria toxin (DTX) transgenic mouse model to specifically target and deplete Treg. METHODS Tumor bearing BALB/c FoxP3.dtr transgenic mice were subjected to different treatment protocols, with or without Treg depletion and tumor growth and survival monitored. RESULTS DTX specifically depleted Treg in a transient, dose-dependent manner. Treg depletion correlated with delayed tumor growth, increased effector T cell (Teff) activation, and enhanced survival in a range of solid tumors. Tumor regression was dependent on Teffs as depletion of both CD4 and CD8 T cells completely abrogated any survival benefit. Severe morbidity following Treg depletion was only observed, when consecutive doses of DTX were given during peak CD8 T cell activation, demonstrating that Treg can be depleted on multiple occasions, but only when CD8 T cell activation has returned to base line levels. Finally, we show that even minimal Treg depletion is sufficient to significantly improve the efficacy of tumor-peptide vaccination. CONCLUSIONS BALB/c.FoxP3.dtr mice are an ideal model to investigate the full therapeutic potential of Treg depletion to boost anti-tumor immunity. DTX-mediated Treg depletion is transient, dose-dependent, and leads to strong anti-tumor immunity and complete tumor regression at high doses, while enhancing the efficacy of tumor-specific vaccination at low doses. Together this data highlight the importance of Treg manipulation as a useful strategy for enhancing current and future cancer immunotherapies.
Collapse
Affiliation(s)
- Scott A. Fisher
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Wayne J. Aston
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Jonathan Chee
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Andrea Khong
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Amanda L. Cleaver
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Jessica N. Solin
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Shaokang Ma
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - W. Joost Lesterhuis
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Ian Dick
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Robert A. Holt
- British Columbia Cancer AgencyVancouverBritish ColumbiaCanada
| | - Jenette Creaney
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | | | - Bruce Robinson
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Richard A. Lake
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| |
Collapse
|
108
|
Putz EM, Guillerey C, Kos K, Stannard K, Miles K, Delconte RB, Takeda K, Nicholson SE, Huntington ND, Smyth MJ. Targeting cytokine signaling checkpoint CIS activates NK cells to protect from tumor initiation and metastasis. Oncoimmunology 2017; 6:e1267892. [PMID: 28344878 DOI: 10.1080/2162402x.2016.1267892] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/22/2016] [Accepted: 11/24/2016] [Indexed: 01/30/2023] Open
Abstract
The cytokine-induced SH2-containing protein CIS belongs to the suppressor of cytokine signaling (SOCS) protein family. Here, we show the critical role of CIS in suppressing natural killer (NK) cell control of tumor initiation and metastasis. Cish-deficient mice were highly resistant to methylcholanthrene-induced sarcoma formation and protected from lung metastasis of B16F10 melanoma and RM-1 prostate carcinoma cells. In contrast, the growth of primary subcutaneous tumors, including those expressing the foreign antigen OVA, was unchanged in Cish-deficient mice. The combination of Cish deficiency and relevant targeted and immuno-therapies such as combined BRAF and MEK inhibitors, immune checkpoint blockade antibodies, IL-2 and type I interferon revealed further improved control of metastasis. The data clearly indicate that targeting CIS promotes NK cell antitumor functions and CIS holds great promise as a novel target in NK cell immunotherapy.
Collapse
Affiliation(s)
- Eva M Putz
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute , Herston, Queensland, Australia
| | - Camille Guillerey
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Kevin Kos
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute , Herston, Queensland, Australia
| | - Kimberley Stannard
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute , Herston, Queensland, Australia
| | - Kim Miles
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute , Herston, Queensland, Australia
| | - Rebecca B Delconte
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Kazuyoshi Takeda
- Department of Immunology, Juntendo University School of Medicine , Bunkyo-ku, Tokyo, Japan
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; School of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
109
|
Ellis JS, Braley-Mullen H. Mechanisms by Which B Cells and Regulatory T Cells Influence Development of Murine Organ-Specific Autoimmune Diseases. J Clin Med 2017; 6:jcm6020013. [PMID: 28134752 PMCID: PMC5332917 DOI: 10.3390/jcm6020013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/21/2016] [Accepted: 01/18/2017] [Indexed: 12/25/2022] Open
Abstract
Experiments with B cell-deficient (B−/−) mice indicate that a number of autoimmune diseases require B cells in addition to T cells for their development. Using B−/− Non-obese diabetic (NOD) and NOD.H-2h4 mice, we demonstrated that development of spontaneous autoimmune thyroiditis (SAT), Sjogren’s syndrome and diabetes do not develop in B−/− mice, whereas all three diseases develop in B cell-positive wild-type (WT) mice. B cells are required early in life, since reconstitution of adult mice with B cells or autoantibodies did not restore their ability to develop disease. B cells function as important antigen presenting cells (APC) to initiate activation of autoreactive CD4+ effector T cells. If B cells are absent or greatly reduced in number, other APC will present the antigen, such that Treg are preferentially activated and effector T cells are not activated. In these situations, B−/− or B cell-depleted mice develop the autoimmune disease when T regulatory cells (Treg) are transiently depleted. This review focuses on how B cells influence Treg activation and function, and briefly considers factors that influence the effectiveness of B cell depletion for treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Jason S Ellis
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA.
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA.
| | - Helen Braley-Mullen
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA.
- Department of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
110
|
Arra A, Lingel H, Kuropka B, Pick J, Schnoeder T, Fischer T, Freund C, Pierau M, Brunner-Weinzierl MC. The differentiation and plasticity of Tc17 cells are regulated by CTLA-4-mediated effects on STATs. Oncoimmunology 2017; 6:e1273300. [PMID: 28344884 DOI: 10.1080/2162402x.2016.1273300] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/08/2016] [Accepted: 12/10/2016] [Indexed: 10/20/2022] Open
Abstract
As the blockade of inhibitory surface-molecules such as CTLA-4 on T cells has led to recent advances in antitumor immune therapy, there is great interest in identifying novel mechanisms of action of CD8+ T cells to evoke effective cytotoxic antitumor responses. Using in vitro and in vivo models, we investigated the molecular pathways underlying the CTLA-4-mediated differentiation of IL-17-producing CD8+ T cells (Tc17 cells) that strongly impairs cytotoxicity. Our studies demonstrate that Tc17 cells lacking CTLA-4 signaling have limited production of STAT3-target gene products such as IL-17, IL-21, IL-23R and RORγt. Upon re-stimulation with IL-12, these cells display fast downregulation of Tc17 hallmarks and acquire Tc1 characteristics such as IFNγ and TNF-α co-expression, which is known to correlate with tumor control. Indeed, upon adoptive transfer, these cells were highly efficient in the antigen-specific rejection of established OVA-expressing B16 melanoma in vivo. Mechanistically, in primary and re-stimulated Tc17 cells, STAT3 binding to the IL-17 promoter was strongly augmented by CTLA-4, associated with less binding of STAT5 and reduced relative activation of STAT1 which is known to block STAT3 activity. Inhibiting CTLA-4-induced STAT3 activity reverses enhancement of signature Tc17 gene products, rendering Tc17 cells susceptible to conversion to Tc1-like cells with enhanced cytotoxic potential. Thus, CTLA-4 critically shapes the characteristics of Tc17 cells by regulating relative STAT3 activation, which provides new perspectives to enhance cytotoxicity of antitumor responses.
Collapse
Affiliation(s)
- Aditya Arra
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Holger Lingel
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Benno Kuropka
- Institut für Chemie und Biochemie, Protein Biochemistry Group, Freie Universität, Berlin, Germany; Mass Spectrometry Group, Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Jonas Pick
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Tina Schnoeder
- Department of Hematology and Oncology, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Thomas Fischer
- Department of Hematology and Oncology, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Christian Freund
- Institut für Chemie und Biochemie, Protein Biochemistry Group, Freie Universität, Berlin, Germany; Mass Spectrometry Group, Leibniz-Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Mandy Pierau
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| | - Monika C Brunner-Weinzierl
- Department of Pediatrics, University Hospital, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University , Magdeburg, Germany
| |
Collapse
|
111
|
Zhang H, Zhang S. The expression of Foxp3 and TLR4 in cervical cancer: association with immune escape and clinical pathology. Arch Gynecol Obstet 2016; 295:705-712. [PMID: 28013346 DOI: 10.1007/s00404-016-4277-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/16/2016] [Indexed: 02/07/2023]
Abstract
PURPOSE To explore the expression of forkhead/winged helix transcription factor p3(Foxp3) and toll-like receptor 4(TLR4) in cervical cancer and evaluate their clinical significance. METHODS Foxp3 and TLR4 protein expression was detected in 105 cervical tissue specimens including cervical cancer, cervical intraepithelial neoplasia (CIN), and healthy control samples using immunohistochemistry. Their relationship with clinicopathologic parameters was also determined. RESULTS Foxp3 and TLR4 had high levels of expression in cervical cancer cells (91.43 and 82.86%, respectively). Foxp3 levels were significantly associated with FIGO stage (P < 0.001) and tumor size (P = 0.034), while TLR4 levels were associated with clinical FIGO stage (P = 0.033) and lymph node metastasis (P = 0.031). Their expression levels were not correlated with age, histologic type, or differentiation (all P > 0.05). These findings suggest that Foxp3 and TLR4 may be useful prognostic indicators of cervical carcinoma. In addition, there were significant positive relationships between Foxp3 and TLR4 expression (r = 0.703, P < 0.001), which shows a possible link and synergistic role of Foxp3 and TLR4 in promoting the immune escape of cervical cancer. CONCLUSIONS Foxp3 and TLR4 may be useful biomarkers for patient prognosis and cervical cancer prediction and treatment.
Collapse
Affiliation(s)
- Huijie Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao St, Heping District, Shenyang, 110004, Liaoning, China
| | - Shulan Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao St, Heping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
112
|
Monzavi-Karbassi B, Jousheghany F, Kieber-Emmons T. Tumor-associated Glycans and Tregs in Immunogenicity of an Autologous Cell-based Vaccine. Immunol Invest 2016; 45:746-758. [PMID: 27759476 DOI: 10.1080/08820139.2016.1219865] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Development of cancer vaccines targeting tumor-associated antigens (TAAs) is an alternative approach to chemotherapy with sustained anti-tumor effects. The success of active immunotherapy has been hampered by tumor-induced immune suppressors. Regulatory T cells (Tregs) are a population of immune suppressors with a proven role in regulating anti-tumor immune responses. Removing or subduing Tregs activity leads to more robust anti-tumor immune responses. Here, we used a cell-based vaccination strategy in the 4T1 murine mammary model to examine whether bulk removal of certain TAAs, using their glycan profile, can affect the immunogenicity of the vaccine. We employed affinity columns of several lectins that are reactive with breast cancer cell lines to deplete lectin-reactive TAAs, while enriching for other antigens. Wheat germ agglutinin (WGA), concanavalin A (Con A), Vicia villosa (VVA), and Griffonia simplicifolia lectin-I (GS-I) were used to fraction crude tumor secreted antigens (TSA). Fractions were tested for their ability to stimulate Tregs and their anti-tumor efficacy. We observed that crude TSA activated Tregs and activation of CD4+CD25+ cells led to an inhibitory function on CD4+CD25- effector cells. Immunization of mice with GS-I- and VVA-depleted fractions significantly delayed tumor establishment and inhibited lung metastases. Depletion of WGA-reactive glycoconjugates led to activation of Tregs, larger tumors and more distant metastases. The data indicate that TAAs can be enriched using their glycan expression pattern to weaken immune suppression and improve anti-tumor response. Therefore, the efficacy of autologous cancer cell vaccination can be improved through enrichment for certain TAAs using carbohydrate specificity.
Collapse
Affiliation(s)
- Behjatolah Monzavi-Karbassi
- a Winthrop P. Rockefeller Cancer Institute and Department of Pathology , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Fariba Jousheghany
- a Winthrop P. Rockefeller Cancer Institute and Department of Pathology , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Thomas Kieber-Emmons
- a Winthrop P. Rockefeller Cancer Institute and Department of Pathology , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| |
Collapse
|
113
|
Abstract
A prerequisite for tumor evolution toward a malignant state is the establishment of cell intrinsic and extrinsic mechanisms of immune suppression (Hanahan and Weinberg, 2000, 2011; Schreiber, Old, and Smyth, 2011). Widespread recruitment of Foxp3+ regulatory T cells (TREG) is a prevailing means to dampen antitumor immunity. Advances in the characterization of TREG cell heterogeneity and physiological function of tissue resident TREG cells unfold new possibilities for nontraditional tumor-promoting functions of intratumoral TREG cells. This review will focus on the nonclassical function of TREG cells and their implicancies for cancer biology and treatment.
Collapse
Affiliation(s)
- Paula D Bos
- a Department of Pathology , Massey Cancer Center, Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|
114
|
Jonuleit H, Bopp T, Becker C. Treg cells as potential cellular targets for functionalized nanoparticles in cancer therapy. Nanomedicine (Lond) 2016; 11:2699-2709. [PMID: 27654070 DOI: 10.2217/nnm-2016-0197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Treg cell-mediated immune suppression appears to represent a significant barrier to effective anticancer immune responses and their inactivation or removal is viewed as a potential therapeutic approach. Although suitable tools for selective Treg cell manipulation in man are missing, their number and function can be altered by a number of drugs and biologicals and by reprogramming tumor-infiltrating antigen presenting cells. Nanoparticles offer exceptional new options in drug and gene delivery by prolonging the circulation time of their cargo, protecting it from degradation and promoting its local accumulation in cells and tissues. In tumor therapy, the use of nanoparticles is expected to overcome limitations in drug delivery and provide novel means for cell-specific functional alteration. In this perspective, we summarize strategies suitable for interference with Treg-mediated suppression, discuss the potential use of nanoparticles for this purpose and identify additional, unexplored opportunities.
Collapse
Affiliation(s)
- Helmut Jonuleit
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| | - Christian Becker
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
115
|
Abstract
The immune system has evolved to defend the organism against an almost infinite number of pathogens in a locally confined and antigen-specific manner while at the same time preserving tolerance to harmless antigens and self. Regulatory T (Treg) cells essentially contribute to an immunoregulatory network preventing excessive immune responses and immunopathology. There is emerging evidence that Treg cells not only operate in secondary lymphoid tissue but also regulate immune responses directly at the site of inflammation. Hence, the classification of Treg cells might need to be further extended by Treg cell subsets that are functionally and phenotypically polarized by their residency. In this review, we discuss recent findings on these tissue-resident Treg cell subsets and how these cells may operate in a tissue- and context-dependent manner.
Collapse
|
116
|
Santegoets SJAM, Welters MJP, van der Burg SH. Monitoring of the Immune Dysfunction in Cancer Patients. Vaccines (Basel) 2016; 4:vaccines4030029. [PMID: 27598210 PMCID: PMC5041023 DOI: 10.3390/vaccines4030029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/25/2016] [Accepted: 08/27/2016] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy shows promising clinical results in patients with different types of cancer, but its full potential is not reached due to immune dysfunction as a result of several suppressive mechanisms that play a role in cancer development and progression. Monitoring of immune dysfunction is a prerequisite for the development of strategies aiming to alleviate cancer-induced immune suppression. At this point, the level at which immune dysfunction occurs has to be established, the underlying mechanism(s) need to be known, as well as the techniques to assess this. While it is relatively easy to measure general signs of immune suppression, it turns out that accurate monitoring of the frequency and function of immune-suppressive cells is still difficult. A lack of truly specific markers, the phenotypic complexity among suppressive cells of the same lineage, but potentially with different functions and functional assays that may not cover every mechanistic aspect of immune suppression are among the reasons complicating proper assessments. Technical innovations in flow and mass cytometry will allow for more complete sets of markers to precisely determine phenotype and associated function. There is, however, a clear need for functional assays that recapitulate more of the mechanisms employed to suppress the immune system.
Collapse
Affiliation(s)
- Saskia J A M Santegoets
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
117
|
Chopra M, Biehl M, Steinfatt T, Brandl A, Kums J, Amich J, Vaeth M, Kuen J, Holtappels R, Podlech J, Mottok A, Kraus S, Jordán-Garrote AL, Bäuerlein CA, Brede C, Ribechini E, Fick A, Seher A, Polz J, Ottmüller KJ, Baker J, Nishikii H, Ritz M, Mattenheimer K, Schwinn S, Winter T, Schäfer V, Krappmann S, Einsele H, Müller TD, Reddehase MJ, Lutz MB, Männel DN, Berberich-Siebelt F, Wajant H, Beilhack A. Exogenous TNFR2 activation protects from acute GvHD via host T reg cell expansion. J Exp Med 2016; 213:1881-900. [PMID: 27526711 PMCID: PMC4995078 DOI: 10.1084/jem.20151563] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/24/2016] [Indexed: 12/22/2022] Open
Abstract
Activation of TNFR2 with a novel agonist expands T reg cells in vivo and protects allo-HCT recipients from acute GvHD while sparing antilymphoma and antiinfectious properties of transplanted donor T cells. Donor CD4+Foxp3+ regulatory T cells (T reg cells) suppress graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (HCT [allo-HCT]). Current clinical study protocols rely on the ex vivo expansion of donor T reg cells and their infusion in high numbers. In this study, we present a novel strategy for inhibiting GvHD that is based on the in vivo expansion of recipient T reg cells before allo-HCT, exploiting the crucial role of tumor necrosis factor receptor 2 (TNFR2) in T reg cell biology. Expanding radiation-resistant host T reg cells in recipient mice using a mouse TNFR2-selective agonist before allo-HCT significantly prolonged survival and reduced GvHD severity in a TNFR2- and T reg cell–dependent manner. The beneficial effects of transplanted T cells against leukemia cells and infectious pathogens remained unaffected. A corresponding human TNFR2-specific agonist expanded human T reg cells in vitro. These observations indicate the potential of our strategy to protect allo-HCT patients from acute GvHD by expanding T reg cells via selective TNFR2 activation in vivo.
Collapse
Affiliation(s)
- Martin Chopra
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Marlene Biehl
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Tim Steinfatt
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Andreas Brandl
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Juliane Kums
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Jorge Amich
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Martin Vaeth
- Department of Molecular Pathology, Institute of Pathology, Würzburg University, 97080 Würzburg, Germany
| | - Janina Kuen
- Department of Molecular Pathology, Institute of Pathology, Würzburg University, 97080 Würzburg, Germany
| | - Rafaela Holtappels
- Institute for Virology and Research Center of Immunotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Jürgen Podlech
- Institute for Virology and Research Center of Immunotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Anja Mottok
- Institute of Pathology, Würzburg University, 97080 Würzburg, Germany
| | - Sabrina Kraus
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Ana-Laura Jordán-Garrote
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Carina A Bäuerlein
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Christian Brede
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Eliana Ribechini
- Institute for Virology and Immunobiology, Würzburg University, 97080 Würzburg, Germany
| | - Andrea Fick
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Axel Seher
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Johannes Polz
- Institute of Immunology, Regensburg University, 93053 Regensburg, Germany
| | - Katja J Ottmüller
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| | - Jeanette Baker
- Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA 94305
| | - Hidekazu Nishikii
- Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA 94305
| | - Miriam Ritz
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Katharina Mattenheimer
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Stefanie Schwinn
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Thorsten Winter
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany
| | - Viktoria Schäfer
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Sven Krappmann
- Microbiology Institute, Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Thomas D Müller
- Department for Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute, Würzburg University, 97080 Würzburg, Germany
| | - Matthias J Reddehase
- Institute for Virology and Research Center of Immunotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, Würzburg University, 97080 Würzburg, Germany
| | - Daniela N Männel
- Institute of Immunology, Regensburg University, 93053 Regensburg, Germany
| | | | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg University, 97080 Würzburg, Germany Center for Interdisciplinary Clinical Research, Würzburg University, 97080 Würzburg, Germany Graduate School of Life Sciences, Würzburg University, 97080 Würzburg, Germany
| |
Collapse
|
118
|
Triner D, Shah YM. Hypoxia-inducible factors: a central link between inflammation and cancer. J Clin Invest 2016; 126:3689-3698. [PMID: 27525434 DOI: 10.1172/jci84430] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The tumor immune response is in a dynamic balance between antitumor mechanisms, which serve to decrease cancer growth, and the protumor inflammatory response, which increases immune tolerance, cell survival, and proliferation. Hypoxia and expression of HIF-1α and HIF-2α are characteristic features of all solid tumors. HIF signaling serves as a major adaptive mechanism in tumor growth in a hypoxic microenvironment. HIFs represent a critical signaling node in the switch to protumorigenic inflammatory responses through recruitment of protumor immune cells and altered immune cell effector functions to suppress antitumor immune responses and promote tumor growth through direct growth-promoting cytokine production, angiogenesis, and ROS production. Modulating HIF function will be an important mechanism to dampen the tumor-promoting inflammatory response and inhibit cancer growth.
Collapse
|
119
|
Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol 2016; 27:1492-504. [PMID: 27207108 DOI: 10.1093/annonc/mdw217] [Citation(s) in RCA: 426] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022] Open
Abstract
The emergence of programmed death-ligand 1 (PD-L1)/programmed death-1 (PD-1)-targeted therapy has demonstrated the importance of the PD-L1 : PD-1 interaction in inhibiting anticancer T-cell immunity in multiple human cancers, generating durable responses and extended overall survival. However, not all patients treated with PD-L1/PD-1-targeted therapy experience tumor shrinkage, durable responses, or prolonged survival. To extend such benefits to more cancer patients, it is necessary to understand why some patients experience primary or secondary immune escape, in which the immune response is incapable of eradicating all cancer cells. Understanding immune escape from PD-L1/PD-1-targeted therapy will be important to the development of rational immune-combination therapy and predictive diagnostics and to the identification of novel immune targets. Factors that likely relate to immune escape include the lack of strong cancer antigens or epitopes recognized by T cells, minimal activation of cancer-specific T cells, poor infiltration of T cells into tumors, downregulation of the major histocompatibility complex on cancer cells, and immunosuppressive factors and cells in the tumor microenvironment. Precisely identifying and understanding these mechanisms of immune escape in individual cancer patients will allow for personalized cancer immunotherapy, in which monotherapy and combination immunotherapy are chosen based on the presence of specific immune biology. This approach may enable treatment with immunotherapy without inducing immune escape, resulting in a larger proportion of patients obtaining clinical benefit.
Collapse
Affiliation(s)
- J M Kim
- Genentech, South San Francisco
| | - D S Chen
- Genentech, South San Francisco Stanford Medical Oncology, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
120
|
Beavis PA, Slaney CY, Kershaw MH, Gyorki D, Neeson PJ, Darcy PK. Reprogramming the tumor microenvironment to enhance adoptive cellular therapy. Semin Immunol 2016; 28:64-72. [DOI: 10.1016/j.smim.2015.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/22/2022]
|
121
|
Abstract
Immunotherapy is now evolving into a major therapeutic option for cancer patients. Such clinical advances also promote massive interest in the search for novel immunotherapy targets, and to understand the mechanism of action of current drugs. It is projected that a series of novel immunotherapy agents will be developed and assessed for their therapeutic activity. In light of this, in vivo experimental mouse models that recapitulate human malignancies serve as valuable tools to validate the efficacy and safety profile of immunotherapy agents, before their transition into clinical trials. In this review, we will discuss the major classes of experimental mouse models of cancer commonly used for immunotherapy assessment and provide examples to guide the selection of appropriate models. We present some new data concerning the utility of a carcinogen-induced tumor model for comparing immunotherapies and combining immunotherapy with chemotherapy. We will also highlight some recent advances in experimental modeling of human malignancies in mice that are leading towards personalized therapy in patients.
Collapse
Affiliation(s)
- Shin Foong Ngiow
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; University of Queensland, Herston, QLD, Australia
| | - Sherene Loi
- Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia; University of Melbourne, Parkville, VIC, Australia
| | - David Thomas
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; University of Queensland, Herston, QLD, Australia; Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia; University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
122
|
Abstract
Unprecedented advances in multiple myeloma (MM) therapy during the last 15 years are predominantly based on our increasing understanding of the pathophysiologic role of the bone marrow (BM) microenvironment. Indeed, new treatment paradigms, which incorporate thalidomide, immunomodulatory drugs (IMiDs), and proteasome inhibitors, target the tumor cell as well as its BM microenvironment. Ongoing translational research aims to understand in more detail how disordered BM-niche functions contribute to MM pathogenesis and to identify additional derived targeting agents. One of the most exciting advances in the field of MM treatment is the emergence of immune therapies including elotuzumab, daratumumab, the immune checkpoint inhibitors, Bispecific T-cell engagers (BiTes), and Chimeric antigen receptor (CAR)-T cells. This chapter will review our knowledge on the pathophysiology of the BM microenvironment and discuss derived novel agents that hold promise to further improve outcome in MM.
Collapse
Affiliation(s)
- Michele Moschetta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yawara Kawano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Klaus Podar
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
123
|
Hahn SA, Bellinghausen I, Trinschek B, Becker C. Translating Treg Therapy in Humanized Mice. Front Immunol 2015; 6:623. [PMID: 26697017 PMCID: PMC4677486 DOI: 10.3389/fimmu.2015.00623] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 12/30/2022] Open
Abstract
Regulatory T cells (Treg) control immune cell function as well as non-immunological processes. Their far-reaching regulatory activities suggest their functional manipulation as a means to sustainably and causally intervene with the course of diseases. Preclinical tools and strategies are however needed to further test and develop interventional strategies outside the human body. “Humanized” mouse models consisting of mice engrafted with human immune cells and tissues provide new tools to analyze human Treg ontogeny, immunobiology, and therapy. Here, we summarize the current state of humanized mouse models as a means to study human Treg function at the molecular level and to design strategies to harness these cells for therapeutic purposes.
Collapse
Affiliation(s)
- Susanne A Hahn
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Iris Bellinghausen
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Bettina Trinschek
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Christian Becker
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| |
Collapse
|
124
|
Castro C, Freitag J, Berod L, Lochner M, Sparwasser T. Microbe-associated immunomodulatory metabolites: Influence on T cell fate and function. Mol Immunol 2015; 68:575-84. [DOI: 10.1016/j.molimm.2015.07.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 06/29/2015] [Accepted: 07/21/2015] [Indexed: 01/30/2023]
|
125
|
The TLR7 agonist induces tumor regression both by promoting CD4⁺T cells proliferation and by reversing T regulatory cell-mediated suppression via dendritic cells. Oncotarget 2015; 6:1779-89. [PMID: 25593198 PMCID: PMC4359331 DOI: 10.18632/oncotarget.2757] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/16/2014] [Indexed: 11/25/2022] Open
Abstract
Treg-induced immunosuppression is now recognized as a key element in enabling tumors to escape immune-mediated destruction. Although topical TLR7 therapies such as imiquimod have been proved successful in the treatment of dermatological malignancy and a number of conditions beyond the FDA-approved indications, the mechanism behind the effect of TLR7 on effector T cell and Treg cell function in cancer immunosurveillance is still not well understood. Here, we found that Loxoribin, one of the TLR7 ligands, could inhibit tumor growth in xenograft models of colon cancer and lung cancer, and these anti-tumor effects of Loxoribin were mediated by promoting CD4⁺T cell proliferation and reversing Treg-mediated suppression via dendritic cells (DCs). However, deprivation of IL-6 using a neutralizing antibody abrogated the ability of Loxoribin-treated DCs, which reversed the Treg cell-mediated suppression. Furthermore, adoptive transfer of Loxoribin-treated DCs inhibited the tumor growth in vivo. Thus, this study links TLR7 signaling to the functional control of effector T cells and Treg cells and identifies Loxoribin as a new therapeutic strategy in cancer treatment, which may offer new opportunities to improve the outcome of cancer immunotherapy.
Collapse
|
126
|
Wang Z, Pratts SG, Zhang H, Spencer PJ, Yu R, Tonsho M, Shah JA, Tanabe T, Powell HR, Huang CA, Madsen JC, Sachs DH, Wang Z. Treg depletion in non-human primates using a novel diphtheria toxin-based anti-human CCR4 immunotoxin. Mol Oncol 2015; 10:553-65. [PMID: 26643572 DOI: 10.1016/j.molonc.2015.11.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 12/15/2022] Open
Abstract
Regulatory T cells (Treg) play an important role in modulating the immune response and has attracted increasing attention in diverse fields such as cancer treatment, transplantation and autoimmune diseases. CC chemokine receptor 4 (CCR4) is expressed on the majority of Tregs, especially on effector Tregs. Recently we have developed a diphtheria-toxin based anti-human CCR4 immunotoxin for depleting CCR4(+) cells in vivo. In this study, we demonstrated that the anti-human CCR4 immunotoxin bound and depleted monkey CCR4(+) cells in vitro. We also demonstrated that the immunotoxin bound to the CCR4(+)Foxp3(+) monkey Tregs in vitro. In vivo studies performed in two naive cynomolgus monkeys revealed 78-89% CCR4(+)Foxp3(+) Treg depletion in peripheral blood lasting approximately 10 days. In lymph nodes, 89-96% CCR4(+)Foxp3(+) Tregs were depleted. No effect was observed in other cell populations including CD8(+) T cells, other CD4(+) T cells, B cells and NK cells. To our knowledge, this is the first agent that effectively depleted non-human primate (NHP) Tregs. This immunotoxin has potential to deplete effector Tregs for combined cancer treatment.
Collapse
Affiliation(s)
- Zhaohui Wang
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shannon G Pratts
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Huiping Zhang
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Philip J Spencer
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruichao Yu
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Makoto Tonsho
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jigesh A Shah
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tatsu Tanabe
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Harrison R Powell
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christene A Huang
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joren C Madsen
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David H Sachs
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; TBRC Laboratories, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhirui Wang
- Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
127
|
Mairhofer DG, Ortner D, Tripp CH, Schaffenrath S, Fleming V, Heger L, Komenda K, Reider D, Dudziak D, Chen S, Becker JC, Flacher V, Stoitzner P. Impaired gp100-Specific CD8(+) T-Cell Responses in the Presence of Myeloid-Derived Suppressor Cells in a Spontaneous Mouse Melanoma Model. J Invest Dermatol 2015; 135:2785-2793. [PMID: 26121214 PMCID: PMC4652066 DOI: 10.1038/jid.2015.241] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 06/01/2015] [Accepted: 06/11/2015] [Indexed: 12/12/2022]
Abstract
Murine tumor models that closely reflect human diseases are important tools to investigate carcinogenesis and tumor immunity. The transgenic (tg) mouse strain tg(Grm1)EPv develops spontaneous melanoma due to ectopic overexpression of the metabotropic glutamate receptor 1 (Grm1) in melanocytes. In the present study, we characterized the immune status and functional properties of immune cells in tumor-bearing mice. Melanoma development was accompanied by a reduction in the percentages of CD4(+) T cells including regulatory T cells (Tregs) in CD45(+) leukocytes present in tumor tissue and draining lymph nodes (LNs). In contrast, the percentages of CD8(+) T cells were unchanged, and these cells showed an activated phenotype in tumor mice. Endogenous melanoma-associated antigen glycoprotein 100 (gp100)-specific CD8(+) T cells were not deleted during tumor development, as revealed by pentamer staining in the skin and draining LNs. They, however, were unresponsive to ex vivo gp100-peptide stimulation in late-stage tumor mice. Interestingly, immunosuppressive myeloid-derived suppressor cells (MDSCs) were recruited to tumor tissue with a preferential accumulation of granulocytic MDSC (grMDSCs) over monocytic MDSC (moMDSCs). Both subsets produced Arginase-1, inducible nitric oxide synthase (iNOS), and transforming growth factor-β and suppressed T-cell proliferation in vitro. In this work, we describe the immune status of a spontaneous melanoma mouse model that provides an interesting tool to develop future immunotherapeutical strategies.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Disease Models, Animal
- Humans
- Lymphocyte Activation
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Random Allocation
- Suppressor Factors, Immunologic/immunology
- Suppressor Factors, Immunologic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Cells, Cultured
- gp100 Melanoma Antigen/immunology
- gp100 Melanoma Antigen/metabolism
Collapse
Affiliation(s)
- David G Mairhofer
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Ortner
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria; Oncotyrol, Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - Sandra Schaffenrath
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria; Oncotyrol, Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - Viktor Fleming
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria; Department of Dermatology, Laboratory of DC-Biology, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Erlangen, Germany
| | - Lukas Heger
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria; Department of Dermatology, Laboratory of DC-Biology, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Erlangen, Germany
| | - Kerstin Komenda
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Reider
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria; Oncotyrol, Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - Diana Dudziak
- Department of Dermatology, Laboratory of DC-Biology, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Erlangen, Germany
| | - Suzie Chen
- Department of Chemical Biology, Lab for Cancer Research, Rutgers University, Piscataway, New Jersey, USA
| | - Jürgen C Becker
- Department for Translational Dermato-Oncology, Center for Medical Biotechnology, University Hospital Essen, Essen, Germany
| | - Vincent Flacher
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
128
|
Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep 2015. [PMID: 26462617 DOI: 10.1038/srep15179.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The prognostic value of FoxP3(+) regulatory T cells (Tregs) in cancer remains controversial. We did a meta-analysis to assess the prognostic effect of FoxP3(+) Treg across different types of cancer and to investigate factors associated with variations in this effect. PubMed, Embase, Cochrane CENTRAL, and Scopus were searched to identify eligible studies. In total, we analyzed 76 articles encompassing 17 types of cancer, and including 15,512 cancer cases. The overall pooled analysis including all types of cancer suggested FoxP3(+)Tregs had a significant negative effect on overall survival (OS) (OR 1.46, P < 0.001), but the prognostic effect varied greatly according to tumor site. High FoxP3(+) Tregs infiltration was significantly associated with shorter OS in the majority of solid tumors studied, including cervical, renal, melanomas, and breast cancers, et al; whereas, FoxP3(+) Tregs were associated with improved survival in colorectal, head and neck, and oesophageal cancers. The stratified analysis suggested the molecular subtype and tumor stage significantly influenced the prognostic value of FoxP3(+) Tregs in certain types of cancer. In conclusion, our meta-analysis suggests that the prognostic role of FoxP3(+) Tregs was highly influenced by tumor site, and was also correlated with the molecular subtype and tumor stage.
Collapse
|
129
|
Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep 2015; 5:15179. [PMID: 26462617 PMCID: PMC4604472 DOI: 10.1038/srep15179] [Citation(s) in RCA: 676] [Impact Index Per Article: 75.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
The prognostic value of FoxP3(+) regulatory T cells (Tregs) in cancer remains controversial. We did a meta-analysis to assess the prognostic effect of FoxP3(+) Treg across different types of cancer and to investigate factors associated with variations in this effect. PubMed, Embase, Cochrane CENTRAL, and Scopus were searched to identify eligible studies. In total, we analyzed 76 articles encompassing 17 types of cancer, and including 15,512 cancer cases. The overall pooled analysis including all types of cancer suggested FoxP3(+)Tregs had a significant negative effect on overall survival (OS) (OR 1.46, P < 0.001), but the prognostic effect varied greatly according to tumor site. High FoxP3(+) Tregs infiltration was significantly associated with shorter OS in the majority of solid tumors studied, including cervical, renal, melanomas, and breast cancers, et al; whereas, FoxP3(+) Tregs were associated with improved survival in colorectal, head and neck, and oesophageal cancers. The stratified analysis suggested the molecular subtype and tumor stage significantly influenced the prognostic value of FoxP3(+) Tregs in certain types of cancer. In conclusion, our meta-analysis suggests that the prognostic role of FoxP3(+) Tregs was highly influenced by tumor site, and was also correlated with the molecular subtype and tumor stage.
Collapse
|
130
|
Kawano Y, Moschetta M, Manier S, Glavey S, Görgün GT, Roccaro AM, Anderson KC, Ghobrial IM. Targeting the bone marrow microenvironment in multiple myeloma. Immunol Rev 2015; 263:160-72. [PMID: 25510276 DOI: 10.1111/imr.12233] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells in the bone marrow (BM). Despite the significant advances in treatment, MM is still a fatal malignancy. This is mainly due to the supportive role of the BM microenvironment in differentiation, migration, proliferation, survival, and drug resistance of the malignant plasma cells. The BM microenvironment is composed of a cellular compartment (stromal cells, osteoblasts, osteoclasts, endothelial cells, and immune cells) and a non-cellular compartment. In this review, we discuss the interaction between the malignant plasma cell and the BM microenvironment and the strategy to target them.
Collapse
Affiliation(s)
- Yawara Kawano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
131
|
Lochner M, Wang Z, Sparwasser T. The Special Relationship in the Development and Function of T Helper 17 and Regulatory T Cells. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 136:99-129. [PMID: 26615094 DOI: 10.1016/bs.pmbts.2015.07.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
T helper 17 (Th17) cells play an essential role in the clearance of extracellular pathogenic bacteria and fungi. However, this subset is critically involved in the pathology of many autoimmune diseases, e.g., psoriasis, multiple sclerosis, allergy, rheumatoid arthritis, and inflammatory bowel diseases in humans. Therefore, Th17 responses need to be tightly regulated in vivo to mediate effective host defenses against pathogens without causing excessive host tissue damage. Foxp3(+) regulatory T (Treg) cells play an important role in maintaining peripheral tolerance to self-antigens and in counteracting the inflammatory activity of effector T helper cell subsets. Although Th17 and Treg cells represent two CD4(+) T cell subsets with opposing principal functions, these cell types are functionally connected. In this review, we will first give an overview on the biology of Th17 cells and describe their development and in vivo function, followed by an account on the special developmental relationship between Th17 and Treg cells. We will describe the identification of Treg/Th17 intermediates and consider their lineage stability and function in vivo. Finally, we will discuss how Treg cells may regulate the Th17 cell response in the context of infection and inflammation, and elude on findings demonstrating that Treg cells can also have a prominent function in promoting the differentiation of Th17 cells.
Collapse
Affiliation(s)
- Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research: A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Zuobai Wang
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research: A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research: A Joint Venture Between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany.
| |
Collapse
|
132
|
Abstract
Chemokines are chemotactic cytokines that control the migration of cells between tissues and the positioning and interactions of cells within tissue. The chemokine superfamily consists of approximately 50 endogenous chemokine ligands and 20 G protein-coupled seven-transmembrane spanning signaling receptors. Chemokines mediate the host response to cancer by directing the trafficking of leukocytes into the tumor microenvironment. This migratory response is complex and consists of diverse leukocyte subsets with both antitumor and protumor activities. Although chemokines were initially appreciated as important mediators of immune cell migration, we now know that they also play important roles in the biology of nonimmune cells important for tumor growth and progression. Chemokines can directly modulate the growth of tumors by inducing the proliferation of cancer cells and preventing their apoptosis. They also direct tumor cell movement required for metastasis. Chemokines can also indirectly modulate tumor growth through their effects on tumor stromal cells and by inducing the release of growth and angiogenic factors from cells in the tumor microenvironment. In this Masters of Immunology primer, we focus on recent advances in understanding the complex nature of the chemokine system in tumor biology with a focus on how the chemokine system could be used to augment cancer immunotherapeutic strategies to elicit a more robust and long-lasting host antitumor immune response.
Collapse
Affiliation(s)
- Melvyn T Chow
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
133
|
Schlößer HA, Theurich S, Shimabukuro-Vornhagen A, Holtick U, Stippel DL, von Bergwelt-Baildon M. Overcoming tumor-mediated immunosuppression. Immunotherapy 2015; 6:973-88. [PMID: 25341119 DOI: 10.2217/imt.14.58] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of tumor-mediated immunosuppression have been described for several solid and hematological tumors. Tumors inhibit immune responses by attraction of immunosuppressive lymphocytic populations, secretion of immunosuppressive cytokines or expression of surface molecules, which inhibit immune responses by induction of anergy or apoptosis in tumor-infiltrating lymphocytes. This tumor-mediated immunosuppression represents a major obstacle to many immunotherapeutic or conventional therapeutic approaches. In this review we discuss how tumor-mediated immunosuppression interferes with different immunotherapeutic approaches and then give an overview of strategies to overcome it. Particular emphasis is placed on agents or approaches already transferred into clinical settings. Finally the success of immune checkpoint inhibitors targeting CTLA-4 or the PD-1 pathway highlights the enormous therapeutic potential of an effective overcoming of tumor-mediated immunosuppression.
Collapse
|
134
|
Kamphorst AO, Araki K, Ahmed R. Beyond adjuvants: immunomodulation strategies to enhance T cell immunity. Vaccine 2015; 33 Suppl 2:B21-8. [PMID: 26022562 PMCID: PMC4449453 DOI: 10.1016/j.vaccine.2014.12.082] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/30/2014] [Accepted: 12/31/2014] [Indexed: 12/31/2022]
Abstract
Engagement of CD8T cells is a crucial aspect of immune responses to pathogens and in tumor surveillance. Nonetheless most vaccination strategies with common adjuvants fail to elicit long-term memory CD8T cells. Increased knowledge on the cellular and molecular requirements for CD8T cell activation has unveiled new opportunities to directly modulate CD8T cells to generate optimal responses. During chronic infections and cancer, immunomodulation strategies to enhance T cell responses may be particularly necessary to overcome the immunosuppressive microenvironment. In this review we will discuss blockade of inhibitory receptors; interleukin-2 administration; regulatory T cell modulation; and targeting of mTOR, as means to enhance CD8T cell immunity.
Collapse
Affiliation(s)
- Alice O Kamphorst
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, 1510 Clifton Rd Rm G211, Atlanta, GA 30322, USA
| | - Koichi Araki
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, 1510 Clifton Rd Rm G211, Atlanta, GA 30322, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, 1510 Clifton Rd Rm G211, Atlanta, GA 30322, USA.
| |
Collapse
|
135
|
Beavis PA, Slaney CY, Kershaw MH, Neeson PJ, Darcy PK. Enhancing the efficacy of adoptive cellular therapy by targeting tumor-induced immunosuppression. Immunotherapy 2015; 7:499-512. [DOI: 10.2217/imt.15.16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Strategies aimed at stimulating the immune system against cancer have signaled a new era for designing new effective therapies for patients. Recent breakthroughs in adoptive cellular therapy and in using checkpoint inhibitors for some patients have renewed much enthusiasm in this field. However, it has become apparent that tumors can use a multitude of inhibitory networks to effectively reduce antitumor immunity. This review discusses our current knowledge of these immune suppressive mechanisms used by tumors and describes potential new strategies that may counteract this problem resulting in significantly increasing therapeutic outcomes of adoptive immunotherapy in a higher proportion of patients.
Collapse
Affiliation(s)
- Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
- Department of Pathology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Clayton, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
- Department of Pathology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Clayton, Australia
| |
Collapse
|
136
|
Cho HI, Jung SH, Sohn HJ, Celis E, Kim TG. An optimized peptide vaccine strategy capable of inducing multivalent CD8 + T cell responses with potent antitumor effects. Oncoimmunology 2015; 4:e1043504. [PMID: 26451316 PMCID: PMC4589052 DOI: 10.1080/2162402x.2015.1043504] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022] Open
Abstract
Therapeutic cancer vaccines are an attractive alternative to conventional therapies for treating malignant tumors, and successful tumor eradication depends primarily on obtaining high numbers of long-lasting tumor-reactive CD8+ T cells. Dendritic cell (DC)-based vaccines constitute a promising approach for treating cancer, but in most instances low immune responses and suboptimal therapeutic effects are achieved indicating that further optimization is required. We describe here a novel vaccination strategy with peptide-loaded DCs followed by a mixture of synthetic peptides, polyinosine-polycytidylic acid (poly-IC) and anti-CD40 antibodies (TriVax) for improving the immunogenicity and therapeutic efficacy of DC-based vaccines in a melanoma mouse model. TriVax immunization 7–12 d after priming with antigen-loaded DCs generated large numbers of long-lasting multiple antigen-specific CD8+ T cells capable of recognizing tumor cells. These responses were far superior to those generated by homologous immunizations with either TriVax or DCs. CD8+ T cells but not CD4+ T cells or NK cells mediated the therapeutic efficacy of this heterologous prime-boost strategy. Moreover, combinations of this vaccination regimen with programmed cell death-1 (PD-1) blockade or IL2 anti-IL2 antibody complexes led to complete disease eradication and survival enhancement in melanoma-bearing mice. The overall results suggest that similar strategies would be applicable for the design of effective therapeutic vaccination for treating viral diseases and various cancers, which may circumvent current limitations of cell-based cancer vaccines.
Collapse
Affiliation(s)
- Hyun-Il Cho
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; Cancer Research Institute; College of Medicine; The Catholic University of Korea ; Seoul, Korea
| | - Soo-Hyun Jung
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; Cancer Research Institute; College of Medicine; The Catholic University of Korea ; Seoul, Korea
| | - Hyun-Jung Sohn
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea
| | - Esteban Celis
- Cancer Immunology; Inflammation and Tolerance Program; Georgia Regents University Cancer Center ; Augusta, GA USA
| | - Tai-Gyu Kim
- Catholic Hematopoietic Stem Cell Bank; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; Cancer Research Institute; College of Medicine; The Catholic University of Korea ; Seoul, Korea ; College of Medicine; The Catholic University of Korea ; Seoul, South Korea
| |
Collapse
|
137
|
Ellis JS, Braley-Mullen H. Regulatory T cells in B-cell-deficient and wild-type mice differ functionally and in expression of cell surface markers. Immunology 2015; 144:598-610. [PMID: 25318356 DOI: 10.1111/imm.12410] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/09/2014] [Accepted: 10/13/2014] [Indexed: 12/13/2022] Open
Abstract
NOD.H-2h4 mice develop spontaneous autoimmune thyroiditis (SAT) with chronic inflammation of thyroids by T and B cells. B-cell deficient (B(-/-) ) mice are resistant to SAT but develop SAT if regulatory T (Treg) cells are transiently depleted. We established a transfer model using splenocytes from CD28(-/-) B(-/-) mice (effector cells and antigen-presenting cells) cultured with or without sorted Treg cells from Foxp3-GFP wild-type (WT) or B(-/-) mice. After transfer to mice lacking T cells, mice given Treg cells from B(-/-) mice had significantly lower SAT severity scores than mice given Treg cells from WT mice, indicating that Treg cells in B(-/-) mice are more effective suppressors of SAT than Treg cells in WT mice. Treg cells from B(-/-) mice differ from WT Treg cells in expression of CD27, tumour necrosis factor receptor (TNFR) II p75, and glucocorticoid-induced TNFR-related protein (GITR). After transient depletion using anti-CD25 or diphtheria toxin, the repopulating Treg cells in B(-/-) mice lack suppressor function, and expression of CD27, GITR and p75 is like that of WT Treg cells. If B(-/-) Treg cells develop with B cells in bone marrow chimeras, their phenotype is like that of WT Treg cells. Addition of B cells to cultures of B(-/-) Treg and T effector cells abrogates their suppressive function and their phenotype is like that of WT Treg cells. These results establish for the first time that Treg cells in WT and B(-/-) mice differ both functionally and in expression of particular cell surface markers. Both properties are altered after transient depletion and repopulation of B(-/-) Treg cells, and by the presence of B cells during Treg cell development or during interaction with effector T cells.
Collapse
Affiliation(s)
- Jason S Ellis
- Department of Medicine, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO, USA
| | | |
Collapse
|
138
|
Klein O, Davis ID, McArthur GA, Chen L, Haydon A, Parente P, Dimopoulos N, Jackson H, Xiao K, Maraskovsky E, Hopkins W, Stan R, Chen W, Cebon J. Low-dose cyclophosphamide enhances antigen-specific CD4(+) T cell responses to NY-ESO-1/ISCOMATRIX™ vaccine in patients with advanced melanoma. Cancer Immunol Immunother 2015; 64:507-18. [PMID: 25662405 PMCID: PMC11029160 DOI: 10.1007/s00262-015-1656-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 01/10/2015] [Indexed: 12/13/2022]
Abstract
Clinical outcomes from cancer vaccine trials in patients with advanced melanoma have so far been disappointing. This appears at least partially due to a state of immunosuppression in these patients induced by an expansion of regulatory cell populations including regulatory T cells (Tregs). We have previously demonstrated potent immunogenicity of the NY-ESO-1/ISCOMATRIX™ vaccine in patients with resected melanoma (study LUD99-08); however, the same vaccine induced only a few vaccine antigen-specific immune responses in patients with advanced disease (study LUD2002-013). Pre-clinical models suggest that the alkylating agent cyclophosphamide can enhance immune responses by depleting Tregs. Therefore, we have enrolled a second cohort of patients with advanced melanoma in the clinical trial LUD2002-013 to investigate whether pre-treatment with cyclophosphamide could improve the immunogenicity of the NY-ESO-1/ISCOMATRIX™ vaccine. The combination treatment led to a significant increase in vaccine-induced NY-ESO-1-specific CD4(+) T cell responses compared with the first trial cohort treated with vaccine alone. We could not detect a significant decline in regulatory T cells in peripheral blood of patients 14 days after cyclophosphamide administration, although a decline at an earlier time point cannot be excluded. Our observations support the inclusion of cyclophosphamide in combination trials with vaccines and other immune-modulatory agents.
Collapse
Affiliation(s)
- Oliver Klein
- Ludwig Institute for Cancer Research (Melbourne-Austin Branch), 147-163 Studley Road, Heidelberg, VIC, 3084, Australia,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
The interplay of effector and regulatory T cells in cancer. Curr Opin Immunol 2015; 33:101-11. [PMID: 25728990 DOI: 10.1016/j.coi.2015.02.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/19/2015] [Accepted: 02/06/2015] [Indexed: 01/05/2023]
Abstract
Regulatory T (Treg) cells suppress effector T (Teff) cells and prevent immune-mediated rejection of cancer. Much less appreciated are mechanisms by which Teff cells antagonize Treg cells. Herein, we consider how complex reciprocal interactions between Teff and Treg cells shape their population dynamics within tumors. Under states of tolerance, including during tumor escape, suppressed Teff cells support Treg cell populations through antigen-dependent provision of interleukin (IL)-2. During immune activation, Teff cells can lose this supportive capacity and directly antagonize Treg cell populations to neutralize their immunosuppressive function. While this latter state is rarely achieved spontaneously within tumors, we propose that therapeutic induction of immune activation has the potential to stably disrupt immunosuppressive population states resulting in durable cancer regression.
Collapse
|
140
|
Workenhe ST, Verschoor ML, Mossman KL. The role of oncolytic virus immunotherapies to subvert cancer immune evasion. Future Oncol 2015; 11:675-89. [DOI: 10.2217/fon.14.254] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
ABSTRACT Despite huge economic and intellectual investments, developing effective cancer treatments continues to be an overarching challenge. Engineered oncolytic viruses (OVs) present self-amplifying immunotherapy platforms capable of preferential cytotoxicity to cancer cells and simultaneous activation of host anti-tumor immunity. In preclinical studies, OVs are showing potent therapeutic effects when used in combination with other immune therapy strategies. In the clinic, the immunotherapeutic effects of OVs are showing promising results. Here we review current strategies for engineering OVs, and present a perspective of future directions within a discussion of the current outcomes of combinatorial approaches with other cancer immunotherapy platforms.
Collapse
Affiliation(s)
- Samuel T Workenhe
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Meghan L Verschoor
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Karen L Mossman
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
141
|
Lees JG, Duffy SS, Perera CJ, Moalem-Taylor G. Depletion of Foxp3+ regulatory T cells increases severity of mechanical allodynia and significantly alters systemic cytokine levels following peripheral nerve injury. Cytokine 2015; 71:207-14. [DOI: 10.1016/j.cyto.2014.10.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/24/2014] [Accepted: 10/28/2014] [Indexed: 11/26/2022]
|
142
|
Increase in Activated Treg in TIL in Lung Cancer and In Vitro Depletion of Treg by ADCC Using an Antihuman CCR4 mAb (KM2760). J Thorac Oncol 2015; 10:74-83. [DOI: 10.1097/jto.0000000000000364] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
143
|
Milanez-Almeida P, Meyer-Hermann M, Toker A, Khailaie S, Huehn J. Foxp3+ regulatory T-cell homeostasis quantitatively differs in murine peripheral lymph nodes and spleen. Eur J Immunol 2014; 45:153-66. [PMID: 25330759 DOI: 10.1002/eji.201444480] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 09/29/2014] [Accepted: 10/17/2014] [Indexed: 11/05/2022]
Abstract
Regulatory T (Treg) cells are essential for maintaining self-tolerance and modulating inflammatory immune responses. Treg cells either develop within the thymus or are converted from CD4(+) naive T (Tnaive) cells in the periphery. The Treg-cell population size is tightly controlled and Treg-cell development and homeostasis have been intensively studied; however, quantitative information about mechanisms of peripheral Treg-cell homeostasis is lacking. Here we developed the first mathematical model of peripheral Treg-cell homeostasis, incorporating secondary lymphoid organs as separate entities and encompassing factors determining the size of the Treg-cell population, namely thymic output, homeostatic proliferation, peripheral conversion, transorgan migration, apoptosis, and the Tnaive-cell population. Quantitative data were collected by monitoring Tnaive-cell homeostasis and Treg-cell rebound after selective in vivo depletion of Treg cells. Our model predicted the previously unanticipated possibility that Treg cells regulate migration of Tnaive cells between spleen and peripheral lymph nodes (LNs), whereas migration of Treg cells between these organs can largely be neglected. Furthermore, our simulations suggested that peripheral conversion significantly contributed to the maintenance of the Treg-cell population, especially in LNs. Hence, we provide the first estimation of the peripheral Treg-cell conversion rate and propose additional facets of Treg-cell-mediated immune regulation that may previously have escaped attention.
Collapse
Affiliation(s)
- Pedro Milanez-Almeida
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | | | | |
Collapse
|
144
|
Lelle M, Hameed A, Ackermann LM, Kaloyanova S, Wagner M, Berisha F, Nikolaev VO, Peneva K. Functional non-nucleoside adenylyl cyclase inhibitors. Chem Biol Drug Des 2014; 85:633-7. [PMID: 25319071 DOI: 10.1111/cbdd.12452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 11/28/2022]
Abstract
In this study, we describe the synthesis of novel functional non-nucleoside adenylyl cyclase inhibitors, which can be easily modified with thiol containing biomolecules such as tumour targeting structures. The linkage between inhibitor and biomolecule contains cleavable bonds to enable efficient intracellular delivery in the reductive milieu of the cytosol as well as in the acidic environment within endosomes and lysosomes. The suitability of this synthetic approach was shown by the successful bioconjugation of a poor cell-permeable inhibitor with a cell-penetrating peptide. Additionally, we have demonstrated the excellent inhibitory effect of the compounds presented here in a live-cell Förster resonance energy transfer-based assay in human embryonic kidney cells.
Collapse
Affiliation(s)
- Marco Lelle
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
145
|
Unger WW, Mayer CT, Engels S, Hesse C, Perdicchio M, Puttur F, Streng-Ouwehand I, Litjens M, Kalay H, Berod L, Sparwasser T, van Kooyk Y. Antigen targeting to dendritic cells combined with transient regulatory T cell inhibition results in long-term tumor regression. Oncoimmunology 2014; 4:e970462. [PMID: 26405564 PMCID: PMC4570108 DOI: 10.4161/21624011.2014.970462] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/20/2022] Open
Abstract
Therapeutic vaccinations against cancer are still largely ineffective. Major caveats are inefficient delivery of tumor antigens to dendritic cells (DCs) and excessive immune suppression by Foxp3+ regulatory T cells (Tregs), resulting in defective T cell priming and failure to induce tumor regression. To circumvent these problems we evaluated a novel combinatorial therapeutic strategy. We show that tumor antigen targeting to DC-SIGN in humanized hSIGN mice via glycans or specific antibodies induces superior T cell priming. Next, this targeted therapy was combined with transient Foxp3+ Treg depletion employing hSIGNxDEREG mice. While Treg depletion alone slightly delayed B16-OVA melanoma growth, only the combination therapy instigated long-term tumor regression in a substantial fraction of mice. This novel strategy resulted in optimal generation of antigen-specific activated CD8+ T cells which accumulated in regressing tumors. Notably, Treg depletion also allowed the local appearance of effector T cells specific for endogenous B16 antigens. This indicates that antitumor immune responses can be broadened by therapies aimed at controlling Tregs in tumor environments. Thus, transient inhibition of Treg-mediated immune suppression potentiates DC targeted antigen vaccination and tumor-specific immunity.
Collapse
Affiliation(s)
- Wendy Wj Unger
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Christian T Mayer
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Steef Engels
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Christina Hesse
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Maurizio Perdicchio
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Franz Puttur
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Ingeborg Streng-Ouwehand
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Manja Litjens
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Luciana Berod
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hanover and the Helmholtz Centre for Infection Research , Hannover, Germany
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology; VU University Medical Center ; Amsterdam, The Netherlands
| |
Collapse
|
146
|
Ngiow SF, von Scheidt B, Möller A, Smyth MJ, Teng MWL. The interaction between murine melanoma and the immune system reveals that prolonged responses predispose for autoimmunity. Oncoimmunology 2014; 2:e23036. [PMID: 23524369 PMCID: PMC3601172 DOI: 10.4161/onci.23036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
An assessment of antitumor immunity versus autoimmunity as provoked by the specific depletion of Foxp3+ Tregs is now possible with the development of Foxp3-diphtheria toxin receptor-like transgenic mouse models. We have used the poorly immunogenic B16F10 melanoma model to characterize a very heterogeneous antitumor effect of the immune response induced by Treg depletion. Depletion and neutralization studies demonstrated the importance of host T cells and interferon γ (IFNγ) in mediating the antitumor response developing in Treg-depleted mice. Such a response correlated with increased proliferation of granzyme B- and IFNγ-producing T cells in the tumor. Furthermore, enhanced antitumor immunity modulated the expression of MHC Class I molecules by B16F10 melanoma cells in Treg-depleted mice. Since Foxp3+ Treg depletion induced a significantly heterogeneous antitumor response, for the first time we were able to assess antitumor immunity and autoimmunity across different groups of responding mice. Strikingly, the duration of the tumor-immune system interaction provoked in individual Treg-depleted mice positively correlated with their propensity to develop vitiligo. A rapid complete tumor rejection was not associated with the development of autoimmunity, however, a proportion of mice that suppressed, but did not effectively clear, B16F10 melanoma did develop vitiligo. The significant implication is that approaches that combine with Treg depletion to rapidly reject tumors may also diminish autoimmune toxicities.
Collapse
Affiliation(s)
- Shin Foong Ngiow
- Cancer Immunology Program; Trescowthick Laboratories; Peter MacCallum Cancer Centre; East Melbourne, Australia ; Department of Pathology; University of Melbourne; Parkville, Australia
| | | | | | | | | |
Collapse
|
147
|
Mayer CT, Lahl K, Milanez-Almeida P, Watts D, Dittmer U, Fyhrquist N, Huehn J, Kopf M, Kretschmer K, Rouse B, Sparwasser T. Advantages of Foxp3(+) regulatory T cell depletion using DEREG mice. IMMUNITY INFLAMMATION AND DISEASE 2014; 2:162-5. [PMID: 25505550 PMCID: PMC4257761 DOI: 10.1002/iid3.33] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/12/2014] [Indexed: 12/17/2022]
Abstract
Several mechanisms enable immunological self-tolerance. Regulatory T cells (Tregs) are a specialized T cell subset that prevents autoimmunity and excessive immune responses, but can also mediate detrimental tolerance to tumors and pathogens in a Foxp3-dependent manner. Genetic tools exploiting the foxp3 locus including bacterial artificial chromosome (BAC)-transgenic DEREG mice have provided essential information on Treg biology and the potential therapeutic modulation of tolerance. In DEREG mice, Foxp3(+) Tregs selectively express eGFP and diphtheria toxin (DT) receptor, allowing for the specific depletion of Tregs through DT administration. We here provide a detailed overview about important considerations such as DT toxicity, which affects any mouse strain treated with DT, and Treg rebound after depletion. Additionally, we point out the specific advantages of BAC-transgenic DEREG mice including their suitability to study organ-specific autoimmunity such as type I diabetes. Moreover, we discuss recent insights into the role of Tregs in viral infections. In summary, DEREG mice are an important tool to study Treg-mediated tolerance and its therapeutic circumvention.
Collapse
Affiliation(s)
- Christian T Mayer
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Katharina Lahl
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine Lane Building, Mailcode 5324, Stanford, CA, 94305, USA ; The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
| | - Pedro Milanez-Almeida
- Experimental Immunology, Helmholtz Centre for Infection Research Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Deepika Watts
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden, Technische Universität Dresden Fetscherstr. 105, 01307, Dresden, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen Virchowstr.179, 45122, Essen, Germany
| | - Nanna Fyhrquist
- Unit of Systems Toxicology, Finnish Institute of Occupational Health Topeliuksenkatu 41b, 00250, Helsinki, Finland
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Manfred Kopf
- Institute for Molecular Health Sciences, Swiss Federal Institute of Technology Zuerich Otto-Stern-Weg 7, 8093, Zuerich, Switzerland
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden, Technische Universität Dresden Fetscherstr. 105, 01307, Dresden, Germany ; Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD) Fetscherstr. 74, 01307, Dresden, Germany
| | - Barry Rouse
- Department of Pathobiology, College of Veterinary Medicine, University of Tennessee Knoxville, TN, 37996, USA
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| |
Collapse
|
148
|
Rosalia RA, Arenas-Ramirez N, Bouchaud G, Raeber ME, Boyman O. Use of enhanced interleukin-2 formulations for improved immunotherapy against cancer. Curr Opin Chem Biol 2014; 23:39-46. [PMID: 25271022 DOI: 10.1016/j.cbpa.2014.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/02/2014] [Accepted: 09/08/2014] [Indexed: 02/02/2023]
Abstract
The use of interleukin-2 (IL-2) for the stimulation of an effector immune response against metastatic cancer dates back to the early 1980s. Administration of unmodified IL-2, either alone or together with antigen-specific approaches, has resulted in remarkably long-term survival of some patients suffering from metastatic melanoma. However, such treatment is usually hampered by the appearance of toxic adverse effects, which has motivated the engineering of modified IL-2 formulations showing reduced toxicity while being more potent at stimulating anti-tumor effector immune cells. In this review we summarize and discuss the features and biological relevance of several enhanced IL-2 formulations, compare these to IL-15-based therapeutics, and try to foreshadow their potential in immunological research and immunotherapy.
Collapse
Affiliation(s)
- Rodney A Rosalia
- Department of Immunology, University Hospital Zurich, Gloriastrasse 30, 8091 Zurich, Switzerland
| | - Natalia Arenas-Ramirez
- Department of Immunology, University Hospital Zurich, Gloriastrasse 30, 8091 Zurich, Switzerland
| | - Grégory Bouchaud
- Institut National de la Recherche Agronomique (INRA), Rue de la Géraudière, BP 71627 Cedex 03, 44316 Nantes, France
| | - Miro E Raeber
- Department of Immunology, University Hospital Zurich, Gloriastrasse 30, 8091 Zurich, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Gloriastrasse 30, 8091 Zurich, Switzerland.
| |
Collapse
|
149
|
Srivastava AK, Dinc G, Sharma RK, Yolcu ES, Zhao H, Shirwan H. SA-4-1BBL and monophosphoryl lipid A constitute an efficacious combination adjuvant for cancer vaccines. Cancer Res 2014; 74:6441-51. [PMID: 25252915 DOI: 10.1158/0008-5472.can-14-1768-a] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Vaccines based on tumor-associated antigens (TAA) have limited therapeutic efficacy due to their weak immunogenic nature and the various immune evasion mechanisms active in advanced tumors. In an effort to overcome these limitations, we evaluated a combination of the T-cell costimulatory molecule SA-4-1BBL with the TLR4 agonist monophosphoryl lipid A (MPL) as a novel vaccine adjuvant system. In the TC-1 mouse allograft model of human papilloma virus (HPV)-induced cancer, a single administration of this combination adjuvant with HPV E7 protein caused tumor rejection in all tumor-bearing mice. On its own, SA-4-1BBL outperformed MPL in this setting. Against established tumors, two vaccinations were sufficient to elicit rejection in the majority of mice. In the metastatic model of Lewis lung carcinoma, vaccination of the TAA survivin with SA-4-1BBL/MPL yielded superior efficacy against pulmonary metastases. Therapeutic efficacy of SA-4-1BBL/MPL was achieved in the absence of detectable toxicity, correlating with enhanced dendritic cell activation, CD8(+) T-cell function, and an increased intratumoral ratio of CD8(+) T effector cells to CD4(+)FoxP3(+) T regulatory cells. Unexpectedly, use of MPL on its own was associated with unfavorable intratumoral ratios of these T-cell populations, resulting in suboptimal efficacy. The efficacy of MPL monotherapy was restored by depletion of T regulatory cells, whereas eliminating CD8(+) T cells abolished the efficacy of its combination with SA-4-1BBL. Mechanistic investigations showed that IFNγ played a critical role in supporting the therapeutic effect of SA-4-1BBL/MPL. Taken together, our results offer a preclinical proof of concept for the use of a powerful new adjuvant system for TAA-based cancer vaccines.
Collapse
Affiliation(s)
- Abhishek K Srivastava
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Gunes Dinc
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Rajesh K Sharma
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Esma S Yolcu
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Hong Zhao
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Haval Shirwan
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
150
|
Ndure J, Flanagan KL. Targeting regulatory T cells to improve vaccine immunogenicity in early life. Front Microbiol 2014; 5:477. [PMID: 25309517 PMCID: PMC4161046 DOI: 10.3389/fmicb.2014.00477] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/25/2014] [Indexed: 12/26/2022] Open
Abstract
Human newborns and infants are bombarded with multiple pathogens on leaving the sterile intra-uterine environment, and yet have suboptimal innate immunity and limited immunological memory, thus leading to increased susceptibility to infections in early life. They are thus the target age group for a host of vaccines against common bacterial and viral pathogens. They are also the target group for many vaccines in development, including those against tuberculosis (TB), malaria, and HIV infection. However, neonatal and infant responses too many vaccines are suboptimal, and in the case of the polysaccharide vaccines, it has been necessary to develop the alternative conjugated formulations in order to induce immunity in early life. Immunoregulatory factors are an intrinsic component of natural immunity necessary to dampen or control immune responses, with the caveat that they may also decrease immunity to infections or lead to chronic infection. This review explores the key immunoregulatory factors at play in early life, with a particular emphasis on regulatory T cells (Tregs). It goes on to explore the role that Tregs play in limiting vaccine immunogenicity, and describes animal and human studies in which Tregs have been depleted in order to enhance vaccine responses. A deeper understanding of the role that Tregs play in limiting or controlling vaccine-induced immunity would provide strategies to improve vaccine immunogenicity in this critical age group. New adjuvants and drugs are being developed that can transiently suppress Treg function, and their use as part of human vaccination strategies against infections is becoming a real prospect for the future.
Collapse
Affiliation(s)
- Jorjoh Ndure
- Infant Immunology Group, Vaccinology Theme, Medical Research Council Laboratories Fajara, The Gambia
| | - Katie L Flanagan
- Vaccine and Infectious Diseases Laboratory, Department of Immunology, Monash University Melbourne, VIC, Australia
| |
Collapse
|