1
|
Blokon-Kogan D, Levi-Mann M, Malka-Levy L, Itzhaki O, Besser MJ, Shiftan Y, Szöőr Á, Vereb G, Gross G, Abken H, Weinstein-Marom H. Membrane anchored IL-18 linked to constitutively active TLR4 and CD40 improves human T cell antitumor capacities for adoptive cell therapy. J Immunother Cancer 2022. [PMCID: PMC9442493 DOI: 10.1136/jitc-2020-001544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BackgroundAdoptive transfer of tumor-infiltrating lymphocytes (TILs) or blood T cells genetically redirected by an antitumor TCR or CAR induces a strong antitumor response in a proportion of patients with cancer; however, the therapeutic efficacy is often limited by rapid decline in T cell functions. Coadministering supportive cytokines frequently provokes systemic side effects preventing their broad clinical application. We recently showed that cytokines can be anchored to the cell membrane in a functional fashion and that cytokine receptor signaling can synergize with TLR4 and CD40 signaling. Here, we aimed at augmenting T cell activation by simultaneous signaling through the cytokine receptor, toll-like receptor and TNF-type receptor using IL-18, TLR4 and CD40 as prototypes.MethodsGenes were expressed on electroporation of in vitro-transcribed mRNA in CD4+ and CD8+ T cells from healthy donors redirected against melanoma cells with an anti-melanotransferrin CAR and in TILs derived from melanoma patients. Functional assays included the activation of signaling pathways, expression of activation and differentiation markers, cytokine secretion and killing of melanoma target cells.ResultsTo provide IL-18 costimulation to T cells in-cis while avoiding systemic effects, we genetically anchored IL-18 to the T cell membrane, either alone (memIL-18) or fused with constitutively active (ca)TLR4 and caCD40 signaling domains arranged in tandem, creating a synthetic ‘all-in-one’ memIL-18-TLR4-CD40 receptor. MemIL-18-TLR4-CD40, but not memIL-18, triggered strong NF-κB activation in cells lacking the IL-18 receptor, attesting to functionality of the TLR-CD40 moiety. While the membrane-anchored cytokine was found to act mainly in-cis, some T cell activation in-trans was also observed. The electroporated T cells exhibited spontaneous T-bet upregulation and IFN-γ and TNF-α secretion. Melanoma-induced activation of CAR-T cells and TILs as manifested by cytokine secretion and cytolytic activity was substantially augmented by both constructs, with memIL-18-TLR4-CD40 exerting stronger effects than memIL-18 alone.ConclusionsLinking membrane anchored IL-18 with caTLR4 and caCD40 signaling in one hybrid transmembrane protein provides simultaneous activation of three T cell costimulatory pathways through one genetically engineered membrane molecule, strongly amplifying T cell functions for adoptive T cell therapy of cancer.
Collapse
Affiliation(s)
- Dayana Blokon-Kogan
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Maya Levi-Mann
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Lior Malka-Levy
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Orit Itzhaki
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center at Tel Hashomer, Ramat Gan, Israel
| | - Michal J Besser
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center at Tel Hashomer, Ramat Gan, Israel
- Department of Clinical Microbiology and Immunology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
- Davidoff Center, Rabin Medical Center, Petah Tikvah, Israel
| | - Yuval Shiftan
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, University of Debrecen Faculty of Medicine, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, University of Debrecen Faculty of Medicine, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, University of Debrecen Faculty of Medicine, Debrecen, Hungary
| | - Gideon Gross
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
| | - Hinrich Abken
- Div. Genetic Immunotherapy, Leibniz Institute for Immunotherapy and University Regensburg, Regensburg, Germany
| | - Hadas Weinstein-Marom
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- The Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee, Israel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center at Tel Hashomer, Ramat Gan, Israel
- Department of Clinical Microbiology and Immunology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
2
|
Rothe F, Patties I, Kortmann RD, Glasow A. Immunomodulatory Effects by Photodynamic Treatment of Glioblastoma Cells In Vitro. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113384. [PMID: 35684322 PMCID: PMC9181863 DOI: 10.3390/molecules27113384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022]
Abstract
Multimodal treatment adding immunotherapy and photodynamic treatment (PDT) to standard therapy might improve the devastating therapeutic outcome of glioblastoma multiforme patients. As a first step, we provide investigations to optimize dendritic cell (DC) vaccination by using PDT and ionizing radiation (IR) to achieve maximal synergistic effects. In vitro experiments were conducted on murine glioblastoma GL261 cells, primary DCs differentiated from bone marrow and T cells, isolated from the spleen. Induction of cell death, reactive oxygen species, and inhibition of proliferation by tetrahydroporphyrin-tetratosylat (THPTS)-PDT and IR were confirmed by WST-1, LDH, ROS, and BrdU assay. Tumor cargo (lysate or cells) for DC load was treated with different combinations of THPTS-PDT, freeze/thaw cycles, and IR and immunogenicity analyzed by induction of T-cell activation. Cellular markers (CD11c, 83, 86, 40, 44, 69, 3, 4, 8, PD-L1) were quantified by flow cytometry. Cytotoxic T-cell response was evaluated by calcein AM assay. Immunogenicity of THPTS-PDT-treated GL261 cells lysate was superior to IR-treated lysate, or treated whole cells proven by increased DC phagocytosis, T-cell adhesion, proliferation, cytolytic activity, and cytokine release. These data strongly support the application of PDT together with IR for optimal immunogenic cell death induction in tumor cell lysate used to pulse DC vaccines.
Collapse
|
3
|
French T, Düsedau HP, Steffen J, Biswas A, Ahmed N, Hartmann S, Schüler T, Schott BH, Dunay IR. Neuronal impairment following chronic Toxoplasma gondii infection is aggravated by intestinal nematode challenge in an IFN-γ-dependent manner. J Neuroinflammation 2019; 16:159. [PMID: 31352901 PMCID: PMC6661741 DOI: 10.1186/s12974-019-1539-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Background It has become increasingly evident that the immune and nervous systems are closely intertwined, relying on one another during regular homeostatic conditions. Prolonged states of imbalance between neural and immune homeostasis, such as chronic neuroinflammation, are associated with a higher risk for neural damage. Toxoplasma gondii is a highly successful neurotropic parasite causing persistent subclinical neuroinflammation, which is associated with psychiatric and neurodegenerative disorders. Little is known, however, by what means neuroinflammation and the associated neural impairment can be modulated by peripheral inflammatory processes. Methods Expression of immune and synapse-associated genes was assessed via quantitative real-time PCR to investigate how T. gondii infection-induced chronic neuroinflammation and associated neuronal alterations can be reshaped by a subsequent acute intestinal nematode co-infection. Immune cell subsets were characterized via flow cytometry in the brain of infected mice. Sulfadiazine and interferon-γ-neutralizing antibody were applied to subdue neuroinflammation. Results Neuroinflammation induced by T. gondii infection of mice was associated with increased microglia activation, recruitment of immune cells into the brain exhibiting Th1 effector functions, and enhanced production of Th1 and pro-inflammatory molecules (IFN-γ, iNOS, IL-12, TNF, IL-6, and IL-1β) following co-infection with Heligmosomoides polygyrus. The accelerated cerebral Th1 immune response resulted in enhanced T. gondii removal but exacerbated the inflammation-related decrease of synapse-associated gene expression. Synaptic proteins EAAT2 and GABAAα1, which are involved in the excitation/inhibition balance in the CNS, were affected in particular. These synaptic alterations were partially recovered by reducing neuroinflammation indirectly via antiparasitic treatment and especially by application of IFN-γ-neutralizing antibody. Impaired iNOS expression following IFN-γ neutralization directly affected EAAT2 and GABAAα1 signaling, thus contributing to the microglial regulation of neurons. Besides, reduced CD36, TREM2, and C1qa gene expression points toward inflammation induced synaptic pruning as a fundamental mechanism. Conclusion Our results suggest that neuroimmune responses following chronic T. gondii infection can be modulated by acute enteric nematode co-infection. While consecutive co-infection promotes parasite elimination in the CNS, it also adversely affects gene expression of synaptic proteins, via an IFN-γ-dependent manner. Electronic supplementary material The online version of this article (10.1186/s12974-019-1539-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Timothy French
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Aindrila Biswas
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Norus Ahmed
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Björn H Schott
- Leibniz Institute of Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Göttingen, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
4
|
Pichler R, Gruenbacher G, Culig Z, Brunner A, Fuchs D, Fritz J, Gander H, Rahm A, Thurnher M. Intratumoral Th2 predisposition combines with an increased Th1 functional phenotype in clinical response to intravesical BCG in bladder cancer. Cancer Immunol Immunother 2016; 66:427-440. [PMID: 28005163 PMCID: PMC5359386 DOI: 10.1007/s00262-016-1945-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/11/2016] [Indexed: 12/11/2022]
Abstract
Th1-type immunity is considered to be required for efficient response to BCG in bladder cancer, although Th2 predisposition of BCG responders has recently been reported. The aim was to evaluate the relationship of Th1 and Th2 components in 23 patients undergoing BCG treatment. Peripheral blood, serum and urine samples were prospectively collected at baseline, during and after BCG. Th1 (neopterin, tryptophan, kynurenine, kynurenine-to-tryptophan ratio (KTR), IL-12, IFN-γ, soluble TNF-R75 and IL-2Rα) and Th2 (IL-4, IL-10) biomarkers as well as CD4 expression in T helper (Th), effector and regulatory T cells were determined. Local immune cell subsets were measured on formalin-fixed, paraffin-embedded cancer tissue by immunohistochemistry to examine expression of transcription factors that control Th1 (T-bet) and Th2-type (GATA3) immunity. We confirmed a Th2 predisposition with a mean GATA3/T-bet ratio of 5.51. BCG responders showed significantly higher levels of urinary (p = 0.003) and serum neopterin (p = 0.012), kynurenine (p = 0.015), KTR (p = 0.005), IFN-γ (p = 0.005) and IL-12 (p = 0.003) during therapy, whereas levels of IL-10 decreased significantly (p < 0.001) compared to non-responders. GATA3/T-bet ratio correlated positively with serum neopterin (p = 0.008), IFN-γ (p = 0.013) and KTR (p = 0.018) after the first BCG instillation. We observed a significant increase in CD4 expression in the Th cell population (p < 0.05), with only a modest tendency toward higher frequency in responders compared to non-responders (p = 0.303). The combined assessment of GATA3/T-bet ratio, neopterin and KTR may be a useful biomarker in predicting BCG response. Th2-promoting factors such as GATA3 may trigger Th1-type immune responses and thus contribute to the BCG success.
Collapse
Affiliation(s)
- Renate Pichler
- Department of Urology, Research Group of Urologic Oncology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Georg Gruenbacher
- Department of Urology, Research Group of Urologic Oncology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.,Immunotherapy Research Unit, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoran Culig
- Department of Urology, Research Group of Urologic Oncology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.,Division of Experimental Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Brunner
- Division of General Pathology, Department of Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Josef Fritz
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Gander
- Department of Urology, Research Group of Urologic Oncology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.,Immunotherapy Research Unit, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Rahm
- Department of Urology, Research Group of Urologic Oncology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.,Immunotherapy Research Unit, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Thurnher
- Department of Urology, Research Group of Urologic Oncology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.,Immunotherapy Research Unit, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
5
|
Nguyen ST, Nguyen HL, Pham VQ, Nguyen GT, Tran CDT, Phan NK, Pham PV. Targeting specificity of dendritic cells on breast cancer stem cells: in vitro and in vivo evaluations. Onco Targets Ther 2015; 8:323-34. [PMID: 25674007 PMCID: PMC4321654 DOI: 10.2147/ott.s77554] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Breast cancer is a leading cause of death in women, and almost all complications are due to chemotherapy resistance. Drug-resistant cells with stem cell phenotypes are thought to cause failure in breast cancer chemotherapy. Dendritic cell (DC) therapy is a potential approach to eradicate these cells. This study evaluates the specificity of DCs for breast cancer stem cells (BCSCs) in vitro and in vivo. BCSCs were enriched by a verapamil-resistant screening method, and reconfirmed by ALDH expression analysis and mammosphere assay. Mesenchymal stem cells (MSCs) were isolated from allogeneic murine bone marrow. DCs were induced from bone marrow-derived monocytes with 20 ng/mL GC-MSF and 20 ng/mL IL-4. Immature DCs were primed with BCSC- or MSC-derived antigens to make two kinds of mature DCs: BCSC-DCs and MSC-DCs, respectively. In vitro ability of BCSC-DCs and MSC-DCs with cytotoxic T lymphocytes (CTLs) to inhibit BCSCs was tested using the xCELLigence technique. In vivo, BCSC-DCs and MSC-DCs were transfused into the peripheral blood of BCSC tumor-bearing mice. The results show that in vitro BCSC-DCs significantly inhibited BCSC proliferation at a DC:CTL ratio of 1:40, while MSC-DCs nonsignificantly decreased BCSC proliferation. In vivo, tumor sizes decreased from 18.8% to 23% in groups treated with BCSC-DCs; in contrast, tumors increased 14% in the control group (RPMI 1640) and 47% in groups treated with MSC-DCs. The results showed that DC therapy could target and be specific to BCSCs. DCs primed with MSCs could trigger tumor growth. These results also indicate that DCs may be a promising therapy for treating drug-resistant cancer cells as well as cancer stem cells.
Collapse
Affiliation(s)
- Sinh Truong Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Huyen Lam Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Viet Quoc Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Giang Thuy Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Cuong Do-Thanh Tran
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Ngoc Kim Phan
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam ; Faculty of Biology, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam ; Faculty of Biology, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| |
Collapse
|
6
|
IL-4: an important cytokine in determining the fate of T cells. Biophys Rev 2014; 6:111-118. [PMID: 28509961 DOI: 10.1007/s12551-013-0133-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 12/03/2013] [Indexed: 02/08/2023] Open
Abstract
The pleiotropic effect of cytokines has been well documented, but the effects triggered by unique cytokines in different T cell types are still under investigation. The most relevant findings on the influence of interleukin-4 (IL-4) on T cell activation, differentiation, proliferation, and survival of different T cell types are discussed in this review. The main aim of our study was to correlate the observed effect with the corresponding molecular mechanism induced on IL-4/IL-4R interaction, in an effort to understand how the same extracellular stimuli can trigger a wide spectrum of signaling pathways leading to different responses in each T cell type.
Collapse
|
7
|
Johnson DJ, Pao LI, Dhanji S, Murakami K, Ohashi PS, Neel BG. Shp1 regulates T cell homeostasis by limiting IL-4 signals. ACTA ACUST UNITED AC 2013; 210:1419-31. [PMID: 23797092 PMCID: PMC3698519 DOI: 10.1084/jem.20122239] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Absence of the phosphatase Shp1 in T cells does not affect the TCR signaling threshold but results in IL-4 sensitivity and memory phenotype cells. The protein-tyrosine phosphatase Shp1 is expressed ubiquitously in hematopoietic cells and is generally viewed as a negative regulatory molecule. Mutations in Ptpn6, which encodes Shp1, result in widespread inflammation and premature death, known as the motheaten (me) phenotype. Previous studies identified Shp1 as a negative regulator of TCR signaling, but the severe systemic inflammation in me mice may have confounded our understanding of Shp1 function in T cell biology. To define the T cell–intrinsic role of Shp1, we characterized mice with a T cell–specific Shp1 deletion (Shp1fl/fl CD4-cre). Surprisingly, thymocyte selection and peripheral TCR sensitivity were unaltered in the absence of Shp1. Instead, Shp1fl/fl CD4-cre mice had increased frequencies of memory phenotype T cells that expressed elevated levels of CD44. Activation of Shp1-deficient CD4+ T cells also resulted in skewing to the Th2 lineage and increased IL-4 production. After IL-4 stimulation of Shp1-deficient T cells, Stat 6 activation was sustained, leading to enhanced Th2 skewing. Accordingly, we observed elevated serum IgE in the steady state. Blocking or genetic deletion of IL-4 in the absence of Shp1 resulted in a marked reduction of the CD44hi population. Therefore, Shp1 is an essential negative regulator of IL-4 signaling in T lymphocytes.
Collapse
Affiliation(s)
- Dylan J Johnson
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C1, Canada
| | | | | | | | | | | |
Collapse
|
8
|
Abstract
CD8(+) T cells play an essential role in immunity against intracellular pathogens, with cytotoxicity being considered their major effector mechanism. However, we here demonstrate that a major part of central and effector memory CD8(+) T cells expresses CD40L, one key molecule for CD4(+) T-cell-mediated help. CD40L(+) CD8(+) T cells are detectable among human antigen-specific immune responses, including pathogens such as influenza and yellow fever virus. CD40L(+) CD8(+) T cells display potent helper functions in vitro and in vivo, such as activation of antigen-presenting cells, and exhibit a cytokine expression signature similar to CD4(+) T cells and unrelated to cytotoxic CD8(+) T cells. The broad occurrence of CD40L(+) CD8(+) T cells in cellular immunity implicates that helper functions are not only executed by major histocompatibility complex (MHC) class II-restricted CD4(+) helper T cells but are also a common feature of MHC class I-restricted CD8(+) T cell responses. Due to their versatile functional capacities, human CD40L(+) CD8(+) T cells are promising candidate cells for immune therapies, particularly when CD4(+) T-cell help or pathogen-associated molecular pattern signals are limited.
Collapse
|
9
|
Kitajima M, Ito T, Tumes DJ, Endo Y, Onodera A, Hashimoto K, Motohashi S, Yamashita M, Nishimura T, Ziegler SF, Nakayama T. Memory type 2 helper T cells induce long-lasting antitumor immunity by activating natural killer cells. Cancer Res 2011; 71:4790-8. [PMID: 21646476 DOI: 10.1158/0008-5472.can-10-1572] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Functionally polarized helper T cells (Th cells) play crucial roles in the induction of tumor immunity. There is considerable knowledge about the contributions of IFN-producing Th1 cells that supports the role of cytotoxic cluster of differentiation (CD8) T cells and natural killer (NK) cells, but much less is known about how IL-4-producing Th2 cells contribute to tumor immunity. In this study, we investigated the cellular and molecular mechanisms employed by memory Th2 cells in sustaining tumor immunity by using a mouse model system wherein ovalbumin (OVA) is used as a specific tumor antigen. In this model, we found that OVA-specific memory Th2 cells exerted potent and long-lasting antitumor effects against NK-sensitive OVA-expressing tumor cells, wherein antitumor effects were mediated by NK cells. Specifically, NK cell cytotoxic activity and expression of perforin and granzyme B were dramatically enhanced by the activation of memory Th2 cells. Interleukin 4 (IL-4) produced by memory Th2 cells in vivo was critical for the antitumor effects of the NK cells, which IL-4 directly stimulated to induce their perforin- and granzyme-B-dependent cytotoxic activity. Our findings show that memory Th2 cells can induce potent antitumor immunity through IL-4-induced activation of NK cells, suggesting potential applications in cellular therapy for cancer patients.
Collapse
Affiliation(s)
- Masayuki Kitajima
- Department of Immunology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Takaku S, Terabe M, Ambrosino E, Peng J, Lonning S, McPherson JM, Berzofsky JA. Blockade of TGF-beta enhances tumor vaccine efficacy mediated by CD8(+) T cells. Int J Cancer 2010; 126:1666-74. [PMID: 19830696 PMCID: PMC2818422 DOI: 10.1002/ijc.24961] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Though TGF-beta inhibition enhances antitumor immunity mediated by CD8(+) T cells in several tumor models, it is not always sufficient for rejection of tumors. In this study, to maximize the antitumor effect of TGF-beta blockade, we tested the effect of anti-TGF-beta combined with an irradiated tumor vaccine in a subcutaneous CT26 colon carcinoma tumor model. The irradiated tumor cell vaccine alone in prophylactic setting significantly delayed tumor growth, whereas anti-TGF-beta antibodies alone did not show any antitumor effect. However, tumor growth was inhibited significantly more in vaccinated mice treated with anti-TGF-beta antibodies compared to vaccinated mice without anti-TGF-beta, suggesting that anti-TGF-beta synergistically enhanced irradiated tumor vaccine efficacy. CD8(+) T-cell depletion completely abrogated the vaccine efficacy, and so protection required CD8(+) T cells. Depletion of CD25(+) T regulatory cells led to the almost complete rejection of tumors without the vaccine, whereas anti-TGF-beta did not change the number of CD25(+) T regulatory cells in unvaccinated and vaccinated mice. Though the abrogation of CD1d-restricted NKT cells, which have been reported to induce TGF-beta production by MDSC through an IL-13-IL-4R-STAT6 pathway, partially enhanced antitumor immunity regardless of vaccination, abrogation of the NKT cell-IL-13-IL-4R-STAT-6 immunoregulatory pathway did not enhance vaccine efficacy. Taken together, these data indicated that anti-TGF-beta enhances efficacy of a prophylactic vaccine in normal individuals despite their not having the elevated TGF-beta levels found in patients with cancer and that the effect is not dependent on TGF-beta solely from CD4(+)CD25(+) T regulatory cells or the NKT cell-IL-13-IL-4R-STAT-6 immunoregulatory pathway.
Collapse
Affiliation(s)
- Shun Takaku
- Vaccine Branch, National Cancer Institute, National Institutes of Health
| | - Masaki Terabe
- Vaccine Branch, National Cancer Institute, National Institutes of Health
| | - Elena Ambrosino
- Vaccine Branch, National Cancer Institute, National Institutes of Health
| | - Judy Peng
- Vaccine Branch, National Cancer Institute, National Institutes of Health
| | | | | | - Jay A. Berzofsky
- Vaccine Branch, National Cancer Institute, National Institutes of Health
| |
Collapse
|
11
|
Kim YS. Tumor Therapy Applying Membrane-bound Form of Cytokines. Immune Netw 2009; 9:158-68. [PMID: 20157604 PMCID: PMC2816950 DOI: 10.4110/in.2009.9.5.158] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 10/11/2009] [Indexed: 12/18/2022] Open
Abstract
Tumor therapy using cytokines has been developed for last two decades. Several recombinant cytokines and tumor cell vaccines produced by cytokine gene transfer have been in clinical trials, but several side effects hamper routine clinical applications. Many cytokines are originally expressed as membrane-bound form and then processed to secretory form exerting paracrine effects. Though functional differences of these two types of cytokines are elusive yet, the membrane-bound form of cytokine may exert its effects on restricted target cells as a juxtacrine, which are in physical contacts. With the efforts to improve antitumor activities of cytokines in cancer patients, developing new strategies to alleviate life-threatening side effects became an inevitable goal of tumor immunologists. Among these, tumor cell vaccines expressing cytokines as membrane-bound form on tumor cell surface have been developed by genetic engineering techniques with the hope of selective stimulation of the target cells that are in cell-to-cell contacts. In this review, recent progress of tumor cell vaccines expressing membrane-bound form of cytokines will be discussed.
Collapse
Affiliation(s)
- Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
12
|
Gil M, Bieniasz M, Wierzbicki A, Bambach BJ, Rokita H, Kozbor D. Targeting a mimotope vaccine to activating Fcgamma receptors empowers dendritic cells to prime specific CD8+ T cell responses in tumor-bearing mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:6808-18. [PMID: 19846865 DOI: 10.4049/jimmunol.0900364] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A major challenge for inducing antitumor immune responses with native or modified tumor/self-Ags in tumor-bearing hosts relates to achieving efficient uptake and processing by dendritic cells (DCs) to activate immune effector cells and limit the generation of regulatory T cell activity. We analyzed the ability of therapeutic DC vaccines expressing a CD166 cross-reactive mimotope of the GD2 ganglioside, 47-LDA, to selectively expand adoptively transferred, tumor-specific T cells in NXS2 neuroblastoma tumor-bearing syngeneic mice. Before the adoptive cell transfer and DC vaccination, the tumor-bearing mice were lymphodepleted by nonmyeloablative total body irradiation or a myeloablative regimen that required bone marrow transplantation. The 47-LDA mimotope was presented to DCs either as a linear polypeptide in conjunction with universal Th epitopes or as a fusion protein with the murine IgG2a Fc fragment (47-LDA-Fcgamma2a) to deliver the antigenic cassette to the activating Fcgamma receptors. We demonstrate that immunization of adoptively transferred T cells in tumor-bearing mice with the 47-LDA mimotope expressed in the context of the activating Fc fusion protein induced higher levels of antitumor immune responses and protection than the 47-LDA polypeptide-DC vaccine. The antitumor efficacy of the therapeutic 47-LDA-Fcgamma2a-DC vaccine was comparable to that achieved by a virotherapy-associated cancer vaccine using a recombinant oncolytic vaccinia virus expressing the 47-LDA-Fcgamma2a fusion protein. The latter treatment, however, did not require total body irradiation or adoptive cell transfer and resulted in induction of antitumor immune responses in the setting of established tolerance, paving the way for testing novel anticancer treatment strategies.
Collapse
Affiliation(s)
- Margaret Gil
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | |
Collapse
|
13
|
Rüder C, Höpken UE, Wolf J, Mittrücker HW, Engels B, Erdmann B, Wollenzin S, Uckert W, Dörken B, Rehm A. The tumor-associated antigen EBAG9 negatively regulates the cytolytic capacity of mouse CD8+ T cells. J Clin Invest 2009; 119:2184-203. [PMID: 19620783 PMCID: PMC2719940 DOI: 10.1172/jci37760] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 05/20/2009] [Indexed: 11/17/2022] Open
Abstract
CTLs eliminate virus-infected and tumorigenic cells through exocytosis of cytotoxic agents from lytic granules. While insights into the intracellular mechanisms facilitating lytic granule release have been obtained through analysis of loss-of-function mutations in humans and mice, there is a paucity of information on negative regulators of secretory lysosome release at the molecular level. By generating and analyzing estrogen receptor-binding fragment-associated antigen 9-KO (Ebag9 KO) mice, we show here that loss of EBAG9 confers CTLs with enhanced cytolytic capacity in vitro and in vivo. Although loss of EBAG9 did not affect lymphocyte development, it led to an increase in CTL secretion of granzyme A, a marker of lytic granules. This resulted in increased cytotoxicity in vitro and an enhanced cytolytic primary and memory T cell response in vivo. We further found that EBAG9 interacts with the adaptor molecule gamma2-adaptin, suggesting EBAG9 is involved in endosomal-lysosomal biogenesis and membrane fusion. Indeed, granzyme B was sorted to secretory lysosomes more efficiently in EBAG9-deficient CTLs than it was in WT CTLs, a finding consistent with the observed enhanced kinetics of cathepsin D proteolytic processing. While EBAG9 deficiency did not disrupt the formation of the immunological synapse, lytic granules in Ebag9-/- CTLs were smaller than in WT CTLs. These data suggest that EBAG9 is a tunable inhibitor of CTL-mediated adaptive immune response functions.
Collapse
Affiliation(s)
- Constantin Rüder
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Uta E. Höpken
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Jana Wolf
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Hans-Willi Mittrücker
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Boris Engels
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Bettina Erdmann
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Susanne Wollenzin
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Wolfgang Uckert
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Bernd Dörken
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| | - Armin Rehm
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
Institute for Immunology, University Medical Center, Hamburg-Eppendorf, Germany.
Universitätsmedizin Berlin, Charité, Virchow Clinics, Berlin, Germany
| |
Collapse
|
14
|
Morris SC, Heidorn SM, Herbert DR, Perkins C, Hildeman DA, Khodoun MV, Finkelman FD. Endogenously produced IL-4 nonredundantly stimulates CD8+ T cell proliferation. THE JOURNAL OF IMMUNOLOGY 2009; 182:1429-38. [PMID: 19155490 DOI: 10.4049/jimmunol.182.3.1429] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell proliferation and survival are regulated by the cytokine receptor common gamma-chain-associated cytokines IL-2, IL-7, and IL-15, while IL-4, another gamma-chain-associated cytokine, is thought to primarily affect T cell quality rather than quantity. In contrast, our experiments reveal that endogenously produced IL-4 is a direct, nonredundant, and potent stimulator of CD8(+) T cell proliferation in Ag- and pathogen-induced CD8(+) T cell responses. These stimulatory effects of IL-4 are observed in both BALB/c and C57BL/6 mice and activate both naive and memory/activated phenotype CD8(+) T cells, although the former are stimulated less than are the latter. IL-4 effects are IL-7- and IL-15-independent, but MHC class I-dependent stimulation appears to be required for the mitogenic effect of IL-4 on naive phenotype CD8(+) T cells. Thus, endogenously produced IL-4 is an important regulator of quantitative as well as qualitative aspects of T cell immunity.
Collapse
Affiliation(s)
- Suzanne C Morris
- Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Muthuswamy R, Urban J, Lee JJ, Reinhart TA, Bartlett D, Kalinski P. Ability of mature dendritic cells to interact with regulatory T cells is imprinted during maturation. Cancer Res 2008; 68:5972-8. [PMID: 18632653 DOI: 10.1158/0008-5472.can-07-6818] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Preferential activation of regulatory T (Treg) cells limits autoimmune tissue damage during chronic immune responses but can also facilitate tumor growth. Here, we show that tissue-produced inflammatory mediators prime maturing dendritic cells (DC) for the differential ability of attracting anti-inflammatory Treg cells. Our data show that prostaglandin E(2) (PGE(2)), a factor overproduced in chronic inflammation and cancer, induces stable Treg-attracting properties in maturing DC, mediated by CCL22. The elevated production of CCL22 by PGE(2)-matured DC persists after the removal of PGE(2) and is further elevated after secondary stimulation of DC in a neutral environment. This PGE(2)-induced overproduction of CCL22 and the resulting attraction of FOXP3(+) Tregs are counteracted by IFN alpha, a mediator of acute inflammation, which also restores the ability of the PGE(2)-exposed DC to secrete the Th1-attracting chemokines: CXCL9, CXCL10, CXCL11, and CCL5. In accordance with these observations, different DCs clinically used as cancer vaccines show different Treg-recruiting abilities, with PGE(2)-matured DC, but not type 1-polarized DC, generated in the presence of type I and type II IFNs, showing high Treg-attracting activity. The current data, showing that the ability of mature DC to interact with Treg cells is predetermined at the stage of DC maturation, pave the way to preferentially target the regulatory versus proinflammatory T cells in autoimmunity and transplantation, as opposed to intracellular infections and cancer.
Collapse
|
16
|
Mempel TR, Bauer CA. Intravital imaging of CD8+ T cell function in cancer. Clin Exp Metastasis 2008; 26:311-27. [PMID: 18665448 DOI: 10.1007/s10585-008-9196-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 07/14/2008] [Indexed: 12/11/2022]
Abstract
Recent technological advances in photonics are making intravital microscopy (IVM) an increasingly powerful approach for the mechanistic exploration of biological processes in the physiological context of complex native tissue environments. Direct, dynamic and multiparametric visualization of immune cell behavior in living animals at cellular and subcellular resolution has already proved its utility in auditing basic immunological concepts established through conventional approaches and has also generated new hypotheses that can conversely be complemented and refined by traditional experimental methods. The insight that outgrowing tumors must not necessarily have evaded recognition by the adaptive immune system, but can escape rejection by actively inducing a state of immunological tolerance calls for a detailed investigation of the cellular and molecular mechanisms by which the anti-cancer response is subverted. Along with molecular imaging techniques that provide dynamic information at the population level, IVM can be expected to make a critical contribution to this effort by allowing the observation of immune cell behavior in vivo at single cell-resolution. We review here how IVM-based investigation can help to clarify the role of cytotoxic T lymphocytes (CTL) in the immune response against cancer and identify the ways by which their function might be impaired through tolerogenic mechanisms.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | | |
Collapse
|
17
|
den Dekker E, Grefte S, Huijs T, ten Dam GB, Versteeg EMM, van den Berk LCJ, Bladergroen BA, van Kuppevelt TH, Figdor CG, Torensma R. Monocyte Cell Surface Glycosaminoglycans Positively Modulate IL-4-Induced Differentiation toward Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:3680-8. [DOI: 10.4049/jimmunol.180.6.3680] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
Landmeier S, Altvater B, Pscherer S, Eing BR, Kuehn J, Rooney CM, Juergens H, Rossig C. Gene-engineered varicella-zoster virus reactive CD4+ cytotoxic T cells exert tumor-specific effector function. Cancer Res 2007; 67:8335-43. [PMID: 17804749 DOI: 10.1158/0008-5472.can-06-4426] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
T cells with grafted specificities for surface antigens provide an avenue for rapidly producing immune effector cells with tumor specificity. However, the function of chimeric receptor (chRec) gene-modified T cells is limited by lack of T-cell expansion and persistence. We propose to use varicella zoster virus (VZV)-reactive T cells as host for the chRec because these cells can be expanded both in vitro and in vivo by stimulation of their native receptor during endogenous reexposure to the virus or by administration of VZV vaccine. We obtained human T cells reactive with VZV from the peripheral blood of seropositive donors by stimulation with VZV lysate and evaluated their characteristics after genetic modification with two tumor-specific model chRecs. Cultures dominated by cytolytic CD4(+) T cells (VZV-CTL) could be expanded and maintained in vitro. Gene-modified VZV-CTL recognized and lysed tumor targets in a MHC-independent manner while maintaining functional, MHC-restricted interaction with VZV antigen through their native receptor. Thus, chRec-transduced VZV-CTL may provide a source of potent tumor-reactive cells for adoptive immunotherapy of cancer. The availability of a safe and effective VZV vaccine provides the option of repeated in vivo stimulation to maintain high T-cell numbers until the tumor is eliminated.
Collapse
MESH Headings
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/physiology
- Cells, Cultured
- Cytotoxicity Tests, Immunologic
- Herpesvirus 3, Human/immunology
- Humans
- Immunotherapy/methods
- Lymphocyte Activation/immunology
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Organisms, Genetically Modified/physiology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Retroviridae/genetics
- Substrate Specificity
- T-Lymphocytes, Cytotoxic/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- Silke Landmeier
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Baschuk N, Utermöhlen O, Gugel R, Warnecke G, Karow U, Paulsen D, Brombacher F, Krönke M, Deppert W. Interleukin-4 impairs granzyme-mediated cytotoxicity of Simian virus 40 large tumor antigen-specific CTL in BALB/c mice. Cancer Immunol Immunother 2007; 56:1625-36. [PMID: 17431618 PMCID: PMC11030854 DOI: 10.1007/s00262-007-0309-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Accepted: 02/26/2007] [Indexed: 11/25/2022]
Abstract
In this report we analyzed the impact of interleukin-4 (IL-4) on tumor-associated simian virus 40 (SV40) large T-antigen (TAg)-specific CD8+ cytotoxic T cells during rejection of syngeneic SV40 transformed mKSA tumor cells in BALB/c mice. Strikingly, challenge of naïve mice with low doses of mKSA tumor cells revealed a CD8+ T cell-dependent prolonged survival time of naïve IL-4-/- mice. In mice immunized with SV40 TAg we observed in IL-4-/- mice, or in wild type mice treated with neutralizing anti-IL-4 monoclonal antibody, a strongly enhanced TAg-specific cytotoxicity of tumor associated CD8+ T cells. The enhanced cytotoxicity in IL-4-/- mice was accompanied by a significant increase in the fraction of CD8+ tumor associated T-cells expressing the cytotoxic effector molecules granzyme A and B and in granzyme B-specific enzymatic activity. The data suggest that endogenous IL-4 can suppress the generation of CD8+ CTL expressing cytotoxic effector molecules especially when the antigen induces only a very weak CTL response.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, Polyomavirus Transforming/immunology
- Antigens, Viral, Tumor/immunology
- Cell Line, Transformed
- Cytotoxicity, Immunologic/genetics
- Granzymes/metabolism
- Interleukin-4/antagonists & inhibitors
- Interleukin-4/genetics
- Interleukin-4/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Mutant Strains
- Neoplasms/immunology
- T-Lymphocytes, Cytotoxic/enzymology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Nikola Baschuk
- Institute for Medical Microbiology, Immunology und Hygiene, Medical Center of the University of Cologne, 50935 Cologne, Germany
| | - Olaf Utermöhlen
- Institute for Medical Microbiology, Immunology und Hygiene, Medical Center of the University of Cologne, 50935 Cologne, Germany
| | - Roland Gugel
- Heinrich-Pette-Institute for Experimental Virology and Immunology, University of Hamburg, 20251 Hamburg, Germany
- Present Address: PolyGene AG, 8153 Rümlang, Switzerland
| | - Gabriele Warnecke
- Heinrich-Pette-Institute for Experimental Virology and Immunology, University of Hamburg, 20251 Hamburg, Germany
| | - Ulrike Karow
- Institute for Medical Microbiology, Immunology und Hygiene, Medical Center of the University of Cologne, 50935 Cologne, Germany
| | - Daniela Paulsen
- Heinrich-Pette-Institute for Experimental Virology and Immunology, University of Hamburg, 20251 Hamburg, Germany
- Present Address: AiCuris GmbH & Co. KG, Aprather Weg 18a / Geb. 405, 42113 Wuppertal, Germany
| | - Frank Brombacher
- Institute for Infectious Diseases and Molecular Medicine (IIDMM), University of Cape Town, 7925 Cape Town, South Africa
| | - Martin Krönke
- Institute for Medical Microbiology, Immunology und Hygiene, Medical Center of the University of Cologne, 50935 Cologne, Germany
- Center for Molecular Medicine, University of Cologne, 50935 Cologne, Germany
| | - Wolfgang Deppert
- Heinrich-Pette-Institute for Experimental Virology and Immunology, University of Hamburg, 20251 Hamburg, Germany
| |
Collapse
|
20
|
De Vleeschouwer S, Spencer Lopes I, Ceuppens JL, Van Gool SW. Persistent IL-10 production is required for glioma growth suppressive activity by Th1-directed effector cells after stimulation with tumor lysate-loaded dendritic cells. J Neurooncol 2007; 84:131-40. [PMID: 17361330 DOI: 10.1007/s11060-007-9362-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Accepted: 02/20/2007] [Indexed: 10/23/2022]
Abstract
Injection of dendritic cells (DC) pulsed with tumor antigens is a novel treatment strategy against malignancies, and aims to elicit anti-tumoral cell-mediated immune responses. We studied the in vitro proliferative responses and cytokine production in T cell cultures after 2 stimulations with autologous DC loaded with tumor lysates derived from glioblastoma multiforme (GBM) cells in the presence of recombinant interleukin (rIL)-6/rIL-12 in the first, and rIL-2/rIL-7 in the second stimulation. After the second stimulation, T cells were co-cultured with glioblastoma (GBM) cells and tumor growth suppression by T cells was assessed using a MTT assay. Although loaded DC induced a significant shift towards T helper cell type 1 (Th1) cytokine production as compared to unloaded DC, persistent interleukin (IL)-10 production by T cells both at the end of 2 stimulations with loaded DC and during the effector phase was also required for their tumor suppressive activity. A stronger glioma growth suppressive activity by T cells stimulated with tumor lysate-loaded DC than by control T cells, cultured with unloaded DC, was seen only if the relative IL-10 production after two stimulations with loaded DC was at least 40% of the IL-10 production after two stimulations with unloaded DC. If less than 40% IL-10 was produced in the experimental condition compared to the control condition, T cells also lost their tumor growth suppressive activity. Addition of rIL-10 during stimulation increased the suppressive activity on tumor cell viability and interferon (IFN)-gamma production by T cells that showed Th1 response upon stimulation with loaded DC. The data point towards the production of both IFN-gamma and IL-10 by responding effector T cells, and towards an immune modulatory rather than immune suppressive role of IL-10 to generate anti-tumoral effector T cells against GBM.
Collapse
Affiliation(s)
- Steven De Vleeschouwer
- Laboratory of Experimental Immunology, Catholic University Leuven, Herestraat 49, Belgium, Leuven 3000, Europe.
| | | | | | | |
Collapse
|
21
|
Boni A, Iezzi G, Degl'Innocenti E, Grioni M, Jachetti E, Camporeale A, Bellone M. Prolonged exposure of dendritic cells to maturation stimuli favors the induction of type-2 cytotoxic T lymphocytes. Eur J Immunol 2006; 36:3157-66. [PMID: 17111344 DOI: 10.1002/eji.200535597] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Dendritic cell (DC) maturation influences the priming and polarization of T lymphocytes. We recently found that early activated DC (i.e. DC exposed to pro-maturation stimuli for 8 h) were more prone to prime in vivo a type-1 cytotoxic T cell (Tc1) response than DC exposed to pro-maturation stimuli for 48 h (48h-DC). We investigated whether 48h-DC, conversely, allowed the induction of Tc2 cells. Antigen-pulsed mouse bone-marrow-derived DC at any maturation stage, in the presence of exogenous IL-12, skewed in vitro naive CD8(+) T cells towards Tc1 cells, but 48h-DC most potently, in the presence of exogenous IL-4, favored the induction of Tc2 cells. In vivo, full maturation of DC promoted expansion of Tc2 and fall of Tc1 cells. Tc2 cells maintained a high cytolytic activity and produced significant amounts of IL-4, IL-5, IL-10 and TGF-beta. Our results indicate that polarization of naive CD8(+) T cells to Tc2 cells is dependent on the amount of time DC have been exposed to maturation stimuli, and might be favored in late and/or chronic phases of an immune response.
Collapse
Affiliation(s)
- Andrea Boni
- Cancer Immunotherapy and Gene Therapy Program, Istituto Scientifico H San Raffaele, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Fröhlich A, Marsland BJ, Sonderegger I, Kurrer M, Hodge MR, Harris NL, Kopf M. IL-21 receptor signaling is integral to the development of Th2 effector responses in vivo. Blood 2006; 109:2023-31. [PMID: 17077330 DOI: 10.1182/blood-2006-05-021600] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Interleukin 21 (IL-21) is a member of the common gamma-chain family of cytokines, which influence a broad spectrum of immunologic responses. A number of studies have examined the function of IL-21, but its specific role in Th1/Th2-cell differentiation and related effector responses remains to be clarified. Thus, we generated IL-21R-deficient mice and have investigated the role of IL-21R signaling using a series of in vivo experimentally induced disease models. We first addressed the role of IL-21R signaling in Th2 immune responses by examining allergic airway inflammation, and Nippostrongylus brasiliensis and Heligmosomoides polygyrus antihelminth responses. In each of these systems, IL-21R signaling played a clear role in the development of Th2 responses. Comparatively, IL-21R signaling was not required for the containment of Leishmania major infection or the development of experimental autoimmune myocarditis, indicative of competent Th1 and Th17 responses, respectively. Adoptive transfer of T cells and analysis of IL-21R+/+/IL-21R-/- chimera mice revealed that IL-21R-signaling was central to Th2-cell survival or migration to peripheral tissues. Overall, our data show IL-21 plays a crucial role in supporting polarized Th2 responses in vivo, while appearing superfluous for Th1 and Th17 responses.
Collapse
Affiliation(s)
- Anja Fröhlich
- Institute of Integrative Biology, Molecular Biomedicine, Swiss Federal Institute of Technology (ETH) Zürich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
23
|
Falkensammer C, Jöhrer K, Gander H, Ramoner R, Putz T, Rahm A, Greil R, Bartsch G, Thurnher M. IL-4 inhibits the TNF-alpha induced proliferation of renal cell carcinoma (RCC) and cooperates with TNF-alpha to induce apoptotic and cytokine responses by RCC: implications for antitumor immune responses. Cancer Immunol Immunother 2006; 55:1228-37. [PMID: 16810557 PMCID: PMC11030668 DOI: 10.1007/s00262-006-0122-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Accepted: 12/28/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVE While previous reports clearly demonstrated antiproliferative effects of IL-4 on renal cell carcinoma (RCC) in vitro, the administration of IL-4 to patients with metastatic RCC in clinical trials could not recapitulate the promising preclinical results. In the present study we wanted to examine the context of IL-4 action and to establish conditions of enhanced IL-4 efficacy. METHODS Primary and permanent human RCC cells were cultured in either serum-supplemented or chemically defined, serum-free culture medium in the presence or absence of cytokines. Cell proliferation was assessed as [(3)H]-thymidine incorporation. Cell apoptosis was measured using the fluorescent DNA intercalator 7-aminoactinomycin D and flow cytometry. In addition, culture media conditioned by RCC were subjected to cytokine antibody array and cytokine multiplex analysis. RESULTS Our results indicate that the previously reported antiproliferative effects of IL-4 are serum-dependent. Under serum-free conditions, IL-4 failed to exhibit growth-inhibitory effects or was even growth-stimulatory. In a chemically defined, serum-free medium (AIM-V), however, IL-4 inhibited the TNF-alpha induced proliferation of RCC. IL-4 and TNF-alpha synergistically induced apoptosis of RCC as well as a complex cytokine response by RCC, which included the synergistic upregulation of RANTES and MCP-1. CONCLUSIONS IL-4 alone has little effect on the spontaneous proliferation of RCC but can prevent the enhancement of proliferation induced by growth promoters like FBS and TNF-alpha. The concomitant growth inhibitory, apoptosis-inducing, and cytokine-enhancing effects of IL-4 in combination with TNF-alpha on RCC support the view that Th2 cytokines may be required for productive immune responses against RCC.
Collapse
Affiliation(s)
- Claudia Falkensammer
- Department of Urology kompetenzzentrum medizin tirol (kmt), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Karin Jöhrer
- Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck, Austria
| | - Hubert Gander
- Department of Urology kompetenzzentrum medizin tirol (kmt), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Reinhold Ramoner
- Department of Urology kompetenzzentrum medizin tirol (kmt), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Thomas Putz
- Department of Urology kompetenzzentrum medizin tirol (kmt), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Andrea Rahm
- Department of Urology kompetenzzentrum medizin tirol (kmt), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Richard Greil
- Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck, Austria
| | - Georg Bartsch
- Department of Urology kompetenzzentrum medizin tirol (kmt), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Martin Thurnher
- Department of Urology kompetenzzentrum medizin tirol (kmt), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
24
|
Casati C, Camisaschi C, Rini F, Arienti F, Rivoltini L, Triebel F, Parmiani G, Castelli C. Soluble human LAG-3 molecule amplifies the in vitro generation of type 1 tumor-specific immunity. Cancer Res 2006; 66:4450-60. [PMID: 16618772 DOI: 10.1158/0008-5472.can-05-2728] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The adjuvant activities of the human lymphocyte activation gene-3 (LAG-3) molecule have been evaluated in a human setting by investigating the ability of a soluble recombinant human LAG-3 protein (hLAG-3Ig) to enhance the in vitro induction of viral- and tumor-specific CTLs. We found that soluble human LAG-3 significantly sustained the generation and expansion of influenza matrix protein Melan-A/MART-1 and survivin-specific CD8+ T lymphocytes in peripheral blood mononuclear cells (PBMC) of both cancer patients and healthy donors, showing its ability to boost CD8+ T-cell memory response or to prime naive T cells in vitro. The peptide-specific T cells generated in the presence of hLAG-3Ig were endowed with cytotoxic activity and enhanced release of type 1 cytotoxic T (Tc1) cytokines and were able to recognize tumor cells expressing their nominal antigen. Phenotype and cytokine/chemokines produced by antigen-presenting cells (APC) of PBMCs exposed in vitro for 2 days to peptide and hLAG-3Ig indicate that the LAG-3-mediated adjuvant effect may depend on a direct activation of circulating APCs. Our data revealed the activity of hLAG-3Ig in inducing tumor-associated, antigen-specific CD8+ T-cell responses in a human setting and strongly support the conclusion that this recombinant protein is a potential candidate adjuvant for cancer vaccines.
Collapse
Affiliation(s)
- Chiara Casati
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Morris SC, Orekhova T, Meadows MJ, Heidorn SM, Yang J, Finkelman FD. IL-4 induces in vivo production of IFN-gamma by NK and NKT cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:5299-305. [PMID: 16621996 DOI: 10.4049/jimmunol.176.9.5299] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Although IL-4 and IFN-gamma often have opposite effects and suppress each other's production by T cells, IL-4 can stimulate IFN-gamma production. To characterize this, we injected mice with IL-4 and quantified IFN-gamma production with the in vivo cytokine capture assay. IL-4 induced Stat6-dependent IFN-gamma production by NK and, to a lesser extent, NKT cells, but not conventional T cells, in 2-4 h. Increased IFN-gamma production persisted at a constant rate for >24 h, but eventually declined, even with continuing IL-4 stimulation. This eventual decline in IFN-gamma production was accompanied by a decrease in NK and T cell numbers. Consistent with a dominant role for NK cells in IL-4-stimulated IFN-gamma secretion, IL-4 induction of IFN-gamma was B and T cell-independent; suppressed by an anti-IL-2Rbeta mAb that eliminates most NK and NKT cells; reduced in Stat4-deficient mice, which have decreased numbers of NK cells; and absent in Rag2/gamma(c)-double-deficient mice, which lack T, B, and NK cells. IL-4-induced IFN-gamma production was not affected by neutralizing IL-12p40 and was increased by neutralizing IL-2. IL-13, which signals through the type 2 IL-4R and mimics many IL-4 effects, failed to stimulate IFN-gamma production and, in most experiments, suppressed basal IFN-gamma production. Thus, IL-4, acting through the type 1 IL-4R, induces Stat6-dependent IFN-gamma secretion by NK and NKT cells. This explains how IL-4 can contribute to Th1 cytokine-associated immune effector functions and suggests how IL-13 can have stronger proallergic effects than IL-4.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Female
- Interferon-gamma/biosynthesis
- Interleukin Receptor Common gamma Subunit
- Interleukin-12/metabolism
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Interleukin-4/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/metabolism
- Male
- Mice
- Mice, Knockout
- Receptors, Interleukin/deficiency
- Receptors, Interleukin/genetics
- Receptors, Interleukin/metabolism
- Receptors, Interleukin-2/immunology
- Receptors, Interleukin-2/metabolism
- STAT4 Transcription Factor/metabolism
- STAT6 Transcription Factor/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Suzanne C Morris
- Division of Immunology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Terabe M, Khanna C, Bose S, Melchionda F, Mendoza A, Mackall CL, Helman LJ, Berzofsky JA. CD1d-Restricted Natural Killer T Cells Can Down-regulate Tumor Immunosurveillance Independent of Interleukin-4 Receptor-Signal Transducer and Activator of Transcription 6 or Transforming Growth Factor-β. Cancer Res 2006; 66:3869-75. [PMID: 16585215 DOI: 10.1158/0008-5472.can-05-3421] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It has been shown previously that the suppression of tumor immunosurveillance may be a mechanism by which tumors resist immune detection and elimination. In this study, we evaluated the role of the immunoregulatory natural killer T (NKT) cells in the biology of immunosurveillance of osteosarcoma. The K7M2 mouse osteosarcoma cell line was implanted orthotopically into wild-type and NKT cell-deficient CD1d knockout (KO) BALB/c mice, and mice were monitored for growth of primary tumors. Further, we examined the role of CD4(+) and/or CD8(+) cells by depleting the cells in vivo and measuring CTL activity in vitro. We also asked the role of interleukin (IL)-4 receptor alpha (IL-4Ralpha)-signal transducer and activator of transcription 6 (STAT6) signaling, including IL-13, and transforming growth factor beta (TGF-beta) by using gene-disrupted mice or treating mice with cytokine antagonists. We were surprised to find a high rate of rejection of osteosarcoma primary tumors in 88% (14 of 16) of CD1d KO mice compared with syngeneic wild-type BALB/c mice that showed rejection of tumor in <24% of mice. Further studies suggested that the rejection of tumor in CD1d KO mice was dependent on CD8(+) lymphocytes. Distinct from other murine tumor models, the negative regulation induced by CD1d-restricted NKT cells was not dependent on IL-4Ralpha-STAT6 signaling, including IL-13, or on TGF-beta. These data suggest that a novel CD1d-restricted NKT cell-mediated mechanism for tumor immunosuppression is active in the K7M2 osteosarcoma model and that NKT cells can regulate immunosurveillance through more than one pathway.
Collapse
Affiliation(s)
- Masaki Terabe
- Vaccine Branch and Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Bldg. 10/Rm. 6B12, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Jarvis LB, Matyszak MK, Duggleby RC, Goodall JC, Hall FC, Gaston JSH. Autoreactive human peripheral blood CD8+ T cells with a regulatory phenotype and function. Eur J Immunol 2005; 35:2896-908. [PMID: 16180249 DOI: 10.1002/eji.200526162] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Despite substantial advances in our understanding of CD4+ CD25+ regulatory T cells, a possible equivalent regulatory subset within the CD8+ T cell population has received less attention. We now describe novel human CD8+/TCR alphabeta+ T cells that have a regulatory phenotype and function. We expanded and cloned these cells using autologous LPS-activated dendritic cells. The clones were not cytolytic, but responded in an autoreactive HLA class I-restricted fashion, by proliferation and production of IL-4, IL-5, IL-13 and TGFbeta1, but not IFN-gamma. They constitutively expressed CD69 and CD25 as well as molecules associated with CD4+ CD25+ regulatory T cells, including cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and Foxp3. They suppressed IFN-gamma production and proliferation by CD4+ T cells in vitro in a cell contact-dependent manner, which could be blocked using a CTLA-4-specific mAb. They were more readily isolated from patients with ankylosing spondylitis and may therefore be up-regulated in response to inflammation. We suggest that they are the CD8+ counterparts of CD4+ CD25+ regulatory T cells. They resemble recently described CD8+ regulatory cells in the rat that were able to abrogate graft-versus-host disease. Likewise, human HLA-restricted CD8+ regulatory T cells that can be cloned and expanded in vitro may have therapeutic applications.
Collapse
Affiliation(s)
- Lorna B Jarvis
- University of Cambridge Clinical School, Dept. of Medicine, Addenbrooke's Hospital, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
28
|
Höpken UE, Lehmann I, Droese J, Lipp M, Schüler T, Rehm A. The ratio between dendritic cells and T cells determines the outcome of their encounter: Proliferationversus deletion. Eur J Immunol 2005; 35:2851-63. [PMID: 16180253 DOI: 10.1002/eji.200526298] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Dendritic cells (DC) either induce T cell tolerance or contribute to the initiation and modulation of T and B cell responses. Since many of the variables determining the thresholds of naive T cell priming were defined in vitro using a homogeneously matured DC population, we here focused on partially mature DC which might reflect the occurrence of tumor-infiltrating and thymic DC. To predict how those DC regulate the induction of antigen-specific T cell proliferation and T cell tolerance, we co-cultured ovalbumin-pulsed murine DC at different ratios with antigen-specific DO11.10 transgenic T cells. Whereas partially mature DC at a DC/T cell ratio of 1:10 supported proliferation, a DC/T cell ratio of 1:2 induced proliferation arrest in naive CD4+ T cells. The acquisition of the NK cell inhibitory markers NK1.1 and KLRG on T cells exposed to high numbers of DC suggests a role for these molecules in the protection of antigen-responsive T cells from exhaustion by overstimulation. Mechanistically, abortive T cell proliferation upon encounter of high numbers of partially mature DC is caused by an apoptosis-related pathway, suggesting that excessive antigen density without sufficient costimulation results in activation-induced cell death.
Collapse
Affiliation(s)
- Uta E Höpken
- Max-Delbrück-Center for Molecular Medicine, Department of Tumorgenetics and Immunogenetics, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Bukreyev A, Belyakov IM, Prince GA, Yim KC, Harris KK, Berzofsky JA, Collins PL. Expression of interleukin-4 by recombinant respiratory syncytial virus is associated with accelerated inflammation and a nonfunctional cytotoxic T-lymphocyte response following primary infection but not following challenge with wild-type virus. J Virol 2005; 79:9515-26. [PMID: 16014914 PMCID: PMC1181599 DOI: 10.1128/jvi.79.15.9515-9526.2005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The outcome of a viral infection or of immunization with a vaccine can be influenced by the local cytokine environment. In studies of experimental vaccines against respiratory syncytial virus (RSV), an increased stimulation of Th2 (T helper 2) lymphocytes was associated with increased immunopathology upon subsequent RSV infection. For this study, we investigated the effect of increased local expression of the Th2 cytokine interleukin-4 (IL-4) from the genome of a recombinant RSV following primary infection and after a challenge with wild-type (wt) RSV. Mice infected with RSV/IL-4 exhibited an accelerated pulmonary inflammatory response compared to those infected with wt RSV, although the wt RSV group caught up by day 8. In the first few days postinfection, RSV/IL-4 was associated with a small but significant acceleration in the expansion of pulmonary T lymphocytes specific for an RSV CD8(+) cytotoxic T-lymphocyte (CTL) epitope presented as a major histocompatibility complex class I tetramer. However, by day 7 the response of tetramer-positive T lymphocytes in the wt RSV group caught up and exceeded that of the RSV/IL-4 group. At all times, the CTL response of the RSV/IL-4 group was deficient in the production of gamma interferon and was nonfunctional for in vitro cell killing. The accelerated inflammatory response coincided with an accelerated accumulation and activation of pulmonary dendritic cells early in infection, but thereafter the dendritic cells were deficient in the expression of B7-1, which governs the acquisition of cytolytic activity by CTL. Following a challenge with wt RSV, there was an increase in Th2 cytokines in the animals that had previously been infected with RSV/IL-4 compared to those previously infected with wt RSV, but the CD8(+) CTL response and the amount of pulmonary inflammation were not significantly different. Thus, a strong Th2 environment during primary pulmonary immunization with live RSV resulted in early inflammation and a largely nonfunctional primary CTL response but had a minimal effect on the secondary response.
Collapse
Affiliation(s)
- Alexander Bukreyev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Cancer Institute, Bethesda, MD 20892-8007, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Eguchi J, Kuwashima N, Hatano M, Nishimura F, Dusak JE, Storkus WJ, Okada H. IL-4-transfected tumor cell vaccines activate tumor-infiltrating dendritic cells and promote type-1 immunity. THE JOURNAL OF IMMUNOLOGY 2005; 174:7194-201. [PMID: 15905564 DOI: 10.4049/jimmunol.174.11.7194] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously demonstrated that IL-4 gene-transfected glioma cell vaccines induce effective therapeutic immunity in preclinical glioma models, and have initiated phase I trials of these vaccines in patients with malignant gliomas. To gain additional mechanistic insight into the efficacy of this approach, we have treated mice bearing the MCA205 (H-2(b)) or CMS-4 (H-2(d)) sarcomas. IL-12/23 p40(-/-) and IFN-gamma(-/-) mice, which were able to reject the initial inoculation of IL-4 expressing tumors, failed to mount a sustained systemic response against parental (nontransfected) tumor cells. Paracrine production of IL-4 in vaccine sites promoted the accumulation and maturation of IL-12p70-secreting tumor-infiltrating dendritic cells (TIDCs). Adoptive transfer of TIDCs isolated from vaccinated wild-type, but not IL-12/23 p40(-/-), mice were capable of promoting tumor-specific CTL responses in syngeneic recipient animals. Interestingly, both STAT4(-/-) and STAT6(-/-) mice failed to reject IL-4-transfected tumors in concert with the reduced capacity of TIDCs to produce IL-12p70 and to promote specific antitumor CTL reactivity. These results suggest that vaccines consisting of tumor cells engineered to produce the type 2 cytokine, IL-4, critically depend on type 1 immunity for their observed therapeutic efficacy.
Collapse
MESH Headings
- Animals
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Movement/immunology
- DNA-Binding Proteins/physiology
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Dendritic Cells/transplantation
- Female
- Fibrosarcoma/genetics
- Fibrosarcoma/immunology
- Fibrosarcoma/pathology
- Graft Rejection/genetics
- Graft Rejection/immunology
- Immunity, Cellular/genetics
- Interferon-gamma/biosynthesis
- Interferon-gamma/physiology
- Interleukin-12/administration & dosage
- Interleukin-12/genetics
- Interleukin-12/physiology
- Interleukin-12 Subunit p40
- Interleukin-4/administration & dosage
- Interleukin-4/biosynthesis
- Interleukin-4/genetics
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Protein Subunits/physiology
- STAT4 Transcription Factor
- STAT6 Transcription Factor
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/pathology
- T-Lymphocyte Subsets/immunology
- Trans-Activators/physiology
- Transfection
Collapse
Affiliation(s)
- Junichi Eguchi
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Gandini S, Lowenfels AB, Jaffee EM, Armstrong TD, Maisonneuve P. Allergies and the risk of pancreatic cancer: a meta-analysis with review of epidemiology and biological mechanisms. Cancer Epidemiol Biomarkers Prev 2005; 14:1908-16. [PMID: 16103436 DOI: 10.1158/1055-9965.epi-05-0119] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Previous reports suggest that allergic disorders may protect against various types of cancer, but the association between history of allergy and pancreatic cancer risk has not been well studied. We did a systematic review and meta-analysis of published studies to evaluate the association of any type, and specific types, of allergy and the risk of pancreatic cancer. We did a comprehensive literature search using MEDLINE, PUBMED, and the ISI Web of Science databases to identify potential relevant case-control and cohort studies. Pooled relative risks (RR) and 95% confidence intervals (95% CI) were calculated using the fixed- and random-effects model. Fourteen population-based studies (4 cohort and 10 case-control studies) with a total of 3,040 pancreatic cancer cases fulfilled our inclusion criteria. A history of allergy was associated with a reduced risk of pancreatic cancer (RR, 0.82; 95% CI, 0.68-0.99). The risk reduction was stronger for allergies related to atopy (RR, 0.71; 95% CI, 0.64-0.80), but not for asthma (RR, 1.01; 95% CI, 0.77-1.31). There was no association between allergies related to food or drugs and pancreatic cancer (RR, 1.08; 95% CI, 0.74-1.58). Overall, there was no evidence of publication bias. Allergies, in particular those related to atopy, seem to be associated with a decreased risk of pancreatic cancer. The hyperactive immune system of allergic individuals may, therefore, in some way lead to increased surveillance and protect against pancreatic cancer development.
Collapse
Affiliation(s)
- Sara Gandini
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | | | | | | | | |
Collapse
|
32
|
Hiraoka SI, Takeuchi N, Bian Y, Nakahara H, Kogo M, Dunussi-Joannopoulos K, Wolf S, Ono S, Fujiwara H. B7.2-Ig fusion proteins enhance IL-4-dependent differentiation of tumor-sensitized CD8+ cytotoxic T lymphocyte precursors. Int Immunol 2005; 17:1071-9. [PMID: 16027141 DOI: 10.1093/intimm/dxh288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The B7/CD28 co-stimulatory pathway plays a critical role in T cell activation and differentiation. Our previous study demonstrated that administration of B7.2-Ig fusion proteins to tumor-bearing mice elicits IL-4-dependent, CD8+ T cell-mediated tumor regression. Here, we investigated whether B7.2-Ig stimulation of tumor-sensitized CD8+ CTL precursors during in vitro antigen re-sensitization actually results in their differentiation into mature CTLs and if so, whether such a process depends on IL-4 signals. Splenocytes from tumor-sensitized (tumor-bearing or tumor-immunized) mice exhibited low levels of anti-tumor CTL responses upon culturing alone, but induced strikingly enhanced CTL responses when stimulated in vitro with B7.2-Ig fusion proteins. Because CTLs were not generated from normal splenocytes even by B7.2-Ig stimulation, the expression of the B7.2-Ig effect required the in vivo tumor sensitization of CD8+ CTL precursors. Administration of anti-CD4 or anti-CD40 ligand (CD40L) to mice before tumor sensitization resulted in almost complete inhibition of CTL responses generated in the subsequent culture containing B7.2-Ig. In contrast, anti-IL-4 did not influence in vivo tumor sensitization required for CTL induction. However, B7.2-Ig stimulation of tumor-sensitized splenocytes enhanced IL-4 production and neutralization of this IL-4 with anti-IL-4 potently down-regulated CTL responses. These results indicate that B7.2-Ig enhances IL-4-dependent differentiation of anti-tumor CD8+ CTL precursors that can be sensitized in vivo depending on collaboration with CD4+ T cells involving CD40L function.
Collapse
Affiliation(s)
- Shin-Ichiro Hiraoka
- Department of Oncology (C6), Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Marsland BJ, Schmitz N, Kopf M. IL-4R? signaling is important for CD8+ T?cell cytotoxicity in the absence of CD4+ T?cell help. Eur J Immunol 2005; 35:1391-8. [PMID: 15816016 DOI: 10.1002/eji.200425768] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cytotoxic CD8(+) T cells are key mediators of viral clearance during primary infection through their production of IFN-gamma and lysis of virally infected cells. Comparatively, the cytokines IL-4 and IL-13 are typically associated with the development of Th2 immune responses against allergens and parasites, while their influence on cytotoxic CD8(+) T cell responses is controversial. We have investigated the roles of IL-4 and IL-13 in the development of CD8(+) T cell responses against influenza infection. We show that in the absence of either IL-4 or IL-13, CD8(+) T cells proliferated and a normal secondary cytotoxic response developed in vitro. In striking contrast, the absence of IL-4Ralpha resulted in impaired ex vivo proliferation and consequently no secondary CTL activity, whereas the in vivo response appeared normal. We show that the presence of CD4(+) T cell help, or the addition of exogenous IL-2 in vitro, restored the response. Taken together, this work reveals previously unrecognized in vivo redundancies between IL-4, IL-13 and IL-2 during immune responses against influenza virus.
Collapse
Affiliation(s)
- Benjamin J Marsland
- Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich-Schlieren, Switzerland
| | | | | |
Collapse
|
34
|
Ishikawa T, Zhu BL, Li DR, Zhao D, Maeda H. Epstein–Barr virus myocarditis as a cause of sudden death: two autopsy cases. Int J Legal Med 2005; 119:231-5. [PMID: 15821942 DOI: 10.1007/s00414-005-0540-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2004] [Accepted: 02/22/2005] [Indexed: 11/24/2022]
Abstract
Although the Epstein-Barr virus (EBV) causes acute infection accompanied by a high fever in young people, there appears to be few reports of a fatal outcome involving myocarditis. We report two cases of unexpected sudden death due to acute myocarditis possibly caused by the EBV. They each visited a hospital due to common cold-like symptoms and unexpectedly died several days later. In both cases, autopsy revealed myocardial necrosis with marked lymphocytic infiltration. Polymerase chain reaction (PCR) screening was positive for the EBV, whereas immunohistochemistry and in situ hybridization for the EBV were negative. Serological investigations showed a mild elevation in antiviral capsid antigen IgG and anti-EBV nuclear antigen IgG in both cases. Immunohistochemical study of lymphocytic infiltrates showed strong positivity for a T-cell marker (CD45R0) in the myocardium and pharyngeal mucosa. These cases suggest the potential risk of mortality from acute EBV infection in young people, even without severe clinical manifestations, and the importance of microbiological investigations, including PCR procedures, in postmortem diagnosis of infectious diseases.
Collapse
Affiliation(s)
- Takaki Ishikawa
- Department of Legal Medicine, Osaka City University Medical School, Asahi-machi 1-4-3, Abeno, Osaka, 545-8558, Japan.
| | | | | | | | | |
Collapse
|
35
|
Abstract
Host protection against a variety of pathogens and tumours requires the efficient induction of CD8(+) T-cell responses. Yet, it has proven difficult to develop vaccines that effectively stimulate this type of cellular immunity. One well-defined obstacle is antigen accessibility to the MHC class I processing pathway. However, cytokines that are produced by cells of the innate immune system also have a key role in CD8(+) T-cell responses, by enhancing 'cross-presentation' and/or inducing CD8(+) T-cell priming and differentiation. Here, we discuss how innate cytokine responses regulate CD8(+) T-cell immunity, and argue that a greater understanding of these processes will be essential for effective tailoring of vaccine-induced cellular immune responses.
Collapse
Affiliation(s)
- Simona Stäger
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| | | |
Collapse
|
36
|
Dobrzanski MJ, Reome JB, Hollenbaugh JA, Dutton RW. Tc1 and Tc2 effector cell therapy elicit long-term tumor immunity by contrasting mechanisms that result in complementary endogenous type 1 antitumor responses. THE JOURNAL OF IMMUNOLOGY 2004; 172:1380-90. [PMID: 14734713 DOI: 10.4049/jimmunol.172.3.1380] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytolytic CD8(+) effector cells fall into two subpopulations based on cytokine secretion. Type 1 CD8(+) T cells (Tc1) secrete IFN-gamma, whereas type 2 CD8(+) T cells (Tc2) secrete IL-4 and IL-5. Both effector cell subpopulations display predominantly perforin-dependent cytolysis in vitro. Using an OVA-transfected B16 lung metastases model, we show that adoptively transferred OVA-specific Tc1 and Tc2 cells induce considerable suppression, but not cure, of pulmonary metastases. However, long-term tumor immunity prolonged survival times indefinitely and was evident by resistance to lethal tumor rechallenge. At early stages after therapy, protection by Tc2 and Tc1 effector cells were dependent in part on effector cell-derived IL-4, IL-5, and IFN-gamma, respectively. Whereas effector cell-derived perforin was not necessary. Over time the numbers of both donor cells diminished to low, yet still detectable, levels. Concomitantly, Tc1 and Tc2 effector cell therapies potentiated endogenous recipient-derived antitumor responses by inducing 1) local T cell-derived chemokines associated with type 1-like immune responses; 2) elevated levels of recipient-derived OVA tetramer-positive CD8 memory T cells that were CD44(high), CD122(+), and Ly6C(high) that predominantly produced IFN-gamma and TNF-alpha; and 3) heightened numbers of activated recipient-derived Th1 and Tc1 T cell subpopulations expressing CD25(+), CD69(+), and CD95(+) cell surface activation markers. Moreover, both Tc2 and Tc1 effector cell therapies were dependent in part on recipient-derived IFN-gamma and TNF-alpha for long-term survival and protection. Collectively, Tc1 and Tc2 effector cell immunotherapy mediate long-term tumor immunity by different mechanisms that subsequently potentiate endogenous recipient-derived type 1 antitumor responses.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Chemokines/biosynthesis
- Chemokines/genetics
- Cytokines/biosynthesis
- Cytokines/metabolism
- Cytokines/physiology
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Immunity, Cellular/genetics
- Immunophenotyping
- Immunotherapy, Adoptive/methods
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Lung/immunology
- Lung/metabolism
- Lung Neoplasms/immunology
- Lung Neoplasms/mortality
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Activation/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/mortality
- Melanoma, Experimental/prevention & control
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasm Transplantation
- Ovalbumin/administration & dosage
- Ovalbumin/genetics
- Ovalbumin/immunology
- Perforin
- Pore Forming Cytotoxic Proteins
- Survival Analysis
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Tumor Necrosis Factor-alpha/deficiency
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/physiology
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
|
37
|
Dobrzanski MJ, Reome JB, Hollenbaugh JA, Hylind JC, Dutton RW. Effector cell-derived lymphotoxin alpha and Fas ligand, but not perforin, promote Tc1 and Tc2 effector cell-mediated tumor therapy in established pulmonary metastases. Cancer Res 2004; 64:406-14. [PMID: 14729652 DOI: 10.1158/0008-5472.can-03-2580] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cytolytic CD8(+) effector cells fall into two subpopulations based on cytokine secretion. Type 1 CD8(+) T cells (Tc1) secrete IFN-gamma, whereas type 2 CD8(+) T cells (Tc2) secrete interleukin (IL)-4 and IL-5. Although both effector cell subpopulations display Fas ligand (FasL) and tumor necrosis factor (TNF), tumor lysis is predominantly perforin dependent in vitro. Using an ovalbumin-transfected B16 lung metastasis model, we show that heightened numbers of adoptively transferred ovalbumin-specific Tc1 and Tc2 cells accumulated at the tumor site by day 2 after therapy and induced tumor regression that enhanced survival in mice with pulmonary metastases. Transfer of either TNF-alpha- or perforin-deficient Tc1 or Tc2 effector cells generated from specified gene-deficient mice showed no differences in therapeutic efficiency when compared with corresponding wild-type cells. In contrast, both Tc1 and Tc2 cells, derived from either FasL or TNF-alpha/lymphotoxin (LT) alpha double knockout mice, showed that therapeutic effects were dependent, in part, on effector cell-derived FasL or LTalpha. Six days after effector cell therapy, elevated levels of activated endogenous CD8/CD44(High) and CD4/CD44(High) T cells localized and persisted at sites of tumor growth, whereas donor cell numbers concomitantly decreased. Both Tc1 and Tc2 effector cell subpopulations induced endogenous antitumor responses that were dependent, in part, on recipient-derived IFN-gamma and TNF-alpha. However, neither effector cell-mediated therapy was dependent on recipient-derived perforin, IL-4, IL-5, or nitric oxide. Collectively, tumor antigen-specific Tc1 and Tc2 effector cell-mediated therapy is initially dependent, in part, on effector cell-derived FasL or LTalpha that may subsequently potentiate endogenous recipient-derived type 1 antitumor responses dependent on TNF-alpha and IFN-gamma.
Collapse
|
38
|
Brunner-Weinzierl MC, Hoff H, Burmester GR. Multiple functions for CD28 and cytotoxic T lymphocyte antigen-4 during different phases of T cell responses: implications for arthritis and autoimmune diseases. Arthritis Res Ther 2004; 6:45-54. [PMID: 15059264 PMCID: PMC400439 DOI: 10.1186/ar1158] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2003] [Revised: 02/11/2004] [Accepted: 02/12/2004] [Indexed: 11/17/2022] Open
Abstract
Chronic T cell responses, as they occur in rheumatoid arthritis, are complex and are likely to involve many mechanisms. There is a growing body of evidence that, in concert with the T cell antigen receptor signal, CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4; CD152) are the primary regulators of T cell responses. Whereas CD28 primarily activates T cell processes, CTLA-4 inhibits them. The mechanism for this dichotomy is not fully understood, especially as CD28 and CTLA-4 recruit similar signalling molecules. In addition, recent studies demonstrate that CD28 and CTLA-4 have multiple functions during T cell responses. In particular, CTLA-4 exerts independent distinct effects during different phases of T cell responses that could be exploited for the treatment of rheumatoid arthritis.
Collapse
|
39
|
Affiliation(s)
- Rémi J Creusot
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, 269 West Campus Drive, Stanford, CA 94305, USA.
| | | |
Collapse
|
40
|
Terabe M, Park JM, Berzofsky JA. Role of IL-13 in regulation of anti-tumor immunity and tumor growth. Cancer Immunol Immunother 2004; 53:79-85. [PMID: 14610620 PMCID: PMC11034335 DOI: 10.1007/s00262-003-0445-0] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2003] [Accepted: 08/07/2003] [Indexed: 11/24/2022]
Abstract
Major mediators of anti-tumor immunity are CD4(+) T(h)1 cells and CD8(+) cytotoxic T lymphocytes (CTLs). In tumor-bearing animals, the T(h)1- and CTL-mediated anti-tumor immunity is down-regulated in multiple ways. Better understanding of negative regulatory pathways of tumor immunity is crucial for the development of anti-tumor vaccines and immunotherapies. Since immune deviation toward T(h)2 suppresses T(h)1 development, it has been thought that induction affecting a T(h)2 immune response is one of the mechanisms that down-regulate effective tumor immune responses. Recent studies using T(h)2-deficient signal transducer and activator (Stat6) KO mice demonstrated that this hypothesis was the case. IL-13 is one of the T(h)2 cytokines that has very similar features to IL-4 through sharing some receptor components and Stat6 signal transduction. It has been thought that IL-13 is not as critical for immune deviation as IL-4 since it cannot directly act on T cells. However, recent studies of IL-13 reveal that this cytokine plays a critical role in many aspects of immune regulation. Studies from our lab and others indicate that IL-13 is central to a novel immunoregulatory pathway in which NKT cells suppress tumor immunosurveillance. Here we will describe biological properties and functions of IL-13, its role in the negative regulation of anti-tumor immunity, and effects of IL-13 on tumor cells themselves.
Collapse
Affiliation(s)
- Masaki Terabe
- Molecular Immunogenetics and Vaccine Research Section, Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1578, USA.
| | | | | |
Collapse
|
41
|
Yamaguchi N, Hiraoka SI, Mukai T, Takeuchi N, Zhou XY, Ono S, Kogo M, Dunussi-Joannopoulos K, Ling V, Wolf S, Fujiwara H. Induction of Tumor Regression by Administration of B7-Ig Fusion Proteins: Mediation by Type 2 CD8+T Cells and Dependence on IL-4 Production. THE JOURNAL OF IMMUNOLOGY 2004; 172:1347-54. [PMID: 14734709 DOI: 10.4049/jimmunol.172.3.1347] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD28 signals contribute to either type 1 or type 2 T cell differentiation. Here, we show that administration of B7.2-Ig fusion proteins to tumor-bearing mice induces tumor regression by promoting the differentiation of antitumor type 2 CD8(+) effector T cells along with IL-4 production. B7.2-Ig-mediated regression was not induced in IL-4(-/-) and STAT6(-/-) mice. However, it was elicited in IFN-gamma(-/-) and STAT4(-/-) mice. By contrast, IL-12-induced tumor regression occurred in IL-4(-/-) and STAT6(-/-) mice, but not in IFN-gamma(-/-) and STAT4(-/-) mice. Moreover, B7.2-Ig treatment was effective in a tumor model not responsive to IL-12. B7.2-Ig administration elicited elevated levels of IL-4 production. Tumor regression was predominantly mediated by CD8(+) T cells, although the induction of these effector cells required CD4(+) T cells. Tumor regression induced by CD8(+) T cells alone was inhibited by neutralizing the IL-4 produced during B7.2-Ig treatment. Thus, these results indicate that stimulation in vivo of CD28 with B7.2-Ig in tumor-bearing mice results in enhanced induction of antitumor type 2 CD8(+) T cells (Tc2) leading to Tc2-mediated tumor regression.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens, CD/administration & dosage
- Antigens, CD/genetics
- Antigens, CD/therapeutic use
- B7-2 Antigen
- CD4-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Female
- Fibrosarcoma/immunology
- Fibrosarcoma/pathology
- Fibrosarcoma/prevention & control
- Immunoglobulin Fc Fragments/administration & dosage
- Immunoglobulin Fc Fragments/genetics
- Immunoglobulin Fc Fragments/therapeutic use
- Inducible T-Cell Co-Stimulator Ligand
- Injections, Subcutaneous
- Interleukin-12/administration & dosage
- Interleukin-4/biosynthesis
- Interleukin-4/deficiency
- Interleukin-4/immunology
- Interleukin-4/physiology
- Male
- Membrane Glycoproteins/administration & dosage
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/therapeutic use
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Proteins/administration & dosage
- Proteins/genetics
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/therapeutic use
- Remission Induction
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Nobuya Yamaguchi
- Department of Oncology, Osaka University Graduate School of Medicine, Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Stäger S, Alexander J, Kirby AC, Botto M, Rooijen NV, Smith DF, Brombacher F, Kaye PM. Natural antibodies and complement are endogenous adjuvants for vaccine-induced CD8+ T-cell responses. Nat Med 2003; 9:1287-92. [PMID: 14502281 DOI: 10.1038/nm933] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2003] [Accepted: 08/26/2003] [Indexed: 12/17/2022]
Abstract
CD8(+) T cells are essential for long-term, vaccine-induced resistance against intracellular pathogens. Here we show that natural antibodies, acting in concert with complement, are endogenous adjuvants for the generation of protective CD8(+) T cells after vaccination against visceral leishmaniasis. IL-4 was crucial for the priming of vaccine-specific CD8(+) T cells, and we defined the primary source of IL-4 as a CD11b(+)CD11c(lo) phagocyte. IL-4 secretion was not observed in antibody-deficient mice and could be reconstituted with serum from normal, but not Btk immune-deficient, mice. Similarly, no IL-4 response or CD8(+) T-cell priming was seen in C1qa(-/-) mice. These results identify a new pathway by which immune complex-mediated complement activation can regulate T-cell-mediated immunity. We propose that this function of natural antibodies could be exploited when developing new vaccines for infectious diseases.
Collapse
Affiliation(s)
- Simona Stäger
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E7HT, UK
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Maszyna F, Hoff H, Kunkel D, Radbruch A, Brunner-Weinzierl MC. Diversity of Clonal T Cell Proliferation Is Mediated by Differential Expression of CD152 (CTLA-4) on the Cell Surface of Activated Individual T Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2003; 171:3459-66. [PMID: 14500641 DOI: 10.4049/jimmunol.171.7.3459] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inhibitory effects of CD152 (CTLA-4) engagement during T cell activation have been described. To date, such effects could only be correlated to CD152 expression at the population level because expression of CD152 on the cell surface is too low to be assessed by conventional immunofluorescence on the single cell level. In this study, we use magnetofluorescent liposomes for the immunofluorescent detection of surface CD152-expressing CD4(+) T cells and show that, despite the fact that nearly all cells express intracellular CD152, only a fraction of 12% of activated T cells expresses surface CD152 at any given time point. Surface CD152(+) T cells appear with similar kinetics after primary or secondary activation in vitro. However, the frequency of surface CD152(+) T cells 48 h postactivation is 2-fold higher during secondary activation. Surface expression of CD152 is independent of the proliferative history of an activated T cell. Instruction of T cells for surface expression of CD152 rather depends on the time elapsed since the onset of activation, with a maximum at 48 h, and requires less than 12 h of Ag exposure. CD152(-) T cells, when isolated by cell sorting and restimulated, continue to proliferate. CD152 blockade has no effect on their proliferation. Isolated surface CD152(+) T cells do not proliferate upon restimulation unless CD152 is blocked. CD152 thus acts directly and autonomously on individual activated and proliferating T lymphocytes. Due to its heterogeneous expression on the cell surface of activated Th cells, CD152 might diversify the T cell response.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigens/pharmacology
- Antigens, CD
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Antigens, Differentiation/physiology
- CTLA-4 Antigen
- Cell Division/immunology
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cell Separation
- Cells, Cultured
- Clone Cells
- Cross-Linking Reagents/metabolism
- Down-Regulation/immunology
- Flow Cytometry
- Immunization, Secondary
- Intracellular Fluid/immunology
- Intracellular Fluid/metabolism
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Time Factors
Collapse
|
44
|
Geginat J, Lanzavecchia A, Sallusto F. Proliferation and differentiation potential of human CD8+ memory T-cell subsets in response to antigen or homeostatic cytokines. Blood 2003; 101:4260-6. [PMID: 12576317 DOI: 10.1182/blood-2002-11-3577] [Citation(s) in RCA: 422] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Four human CD8+ T-cell subsets, naive (CCR7+CD45RA+), central memory (TCM, CCR7+CD45RA-), effector memory (TEM, CCR7-CD45RA-), and CD45RA+ effector memory cells (TEMRA, CCR7-CD45RA+) were compared for their capacity to proliferate and differentiate in response to antigen or homeostatic cytokines. Cytokine responsiveness and interleukin-15 receptor expression were low in naive T cells and progressively increased from TCM to TEM and TEMRA. In contrast, the capacity to accumulate in response to T-cell receptor (TCR) or cytokine stimulation showed a reciprocal pattern and was associated with resistance to cell death and Bcl-2 expression. Whereas all TCR-stimulated cells acquired a CD45RA-CCR7- phenotype, cytokine-stimulated cells maintained their phenotype with the exception of TCM cells, which expressed CCR7, CD45RA, and perforin in various combinations. Single CD8+ TCM cells, but not TEM cells, could be expanded with cytokines, and the obtained clones displayed several distinct phenotypes, suggesting that TCM cells are heterogeneous. Consistently, CCR4 expression in the CD8+ TCM pool discriminated CCR4+ type 2 polarized cells (Tc2) and CCR4-CTL precursors. Finally, ex vivo bromodeoxyuridine (BrdU) incorporation experiments revealed that memory subsets have different in vivo proliferation rates, with CCR4-TCM having the highest turnover and TEMRA the lowest. These results show that human CD8+ memory T-cell subsets have different proliferation and differentiation potentials in vitro and in vivo. Furthermore, they suggest that TEMRA cells are generated from a TCM subset upon homeostatic proliferation in the absence of antigen.
Collapse
Affiliation(s)
- Jens Geginat
- Institute for Research in Biomedicine, Bellinzona, Switzerland.
| | | | | |
Collapse
|
45
|
Blankenstein T, Qin Z. The role of IFN-gamma in tumor transplantation immunity and inhibition of chemical carcinogenesis. Curr Opin Immunol 2003; 15:148-54. [PMID: 12633663 DOI: 10.1016/s0952-7915(03)00007-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
IFN-gamma contributes to the rejection of transplantable tumors and the inhibition of methylcholanthrene (MCA)-induced carcinogenesis by different mechanisms. In most tumor transplantation models, tumor rejection requires IFN-gamma receptor expression by host cells, but not by tumor cells. IFN-gamma produced by either CD4+ or CD8+ T cells acts on non-hematopoietic tumor stroma cells and, either directly or indirectly, induces angiostasis. This prevents rapid tumor burden and allows residual tumor cells to be eliminated. In some models, IFN-gamma also contributes to the destruction of existing tumor blood vessels. During MCA-induced tumorigenesis IFN-gamma is involved in the inhibition of MCA diffusion by encapsulation and reduction of DNA damage. This mechanism may primarily protect tissue from damage and simultaneously inhibit tumor development.
Collapse
|
46
|
Gey A, Kumari P, Sambandam A, Lecuru F, Cassard L, Badoual C, Fridman C, Nagarajan B, Fridman WH, Tartour E. Identification and characterisation of a group of cervical carcinoma patients with profound downregulation of intratumoral Type 1 (IFNgamma) and Type 2 (IL-4) cytokine mRNA expression. Eur J Cancer 2003; 39:595-603. [PMID: 12628838 DOI: 10.1016/s0959-8049(02)00839-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Type 1 cytokines, such as interferon gamma (IFNgamma) and interleukin-2 (IL-2), increase T cell-mediated immune responses and are considered to be beneficial for antitumour immunity. Type 2 cytokines, such as IL-4, IL-5, and IL-10, inhibit Type 1 responses and promote humoral responses. We have previously reported an association between low intratumoral IFNgamma mRNA levels and poor clinical outcome in patients with invasive cervical carcinoma. In this study, by using quantitative polymerase chain reaction (PCR), we identified a group of cervical carcinoma patients with undetectable intratumoral T cell-derived cytokine mRNAs, as IFNgamma, IL-4 and IL-17 expression could not be detected in 5, 25 and 8 of the 52 biopsies analysed, respectively. Global downregulation of Type 1 and Type 2 cytokines was observed in a subgroup of patients who more frequently presented advanced stage tumours. Biopsies of patients with no IFNgamma gene expression did not appear to be less infiltrated by T cells than control biopsies with measurable IFNgamma gene expression. These results clearly demonstrate that, in some clinical situations, the decrease in intratumoral Type 1 cytokines is not associated with a Type 2 polarisation, but rather reflects global deactivation of T cells at the tumour site. These data provide support for immunotherapy protocols designed to reverse the anergic state of T cells in cancer.
Collapse
Affiliation(s)
- A Gey
- Unité d'immunologie biologique, Hopital Européen Georges Pompidou, INSERM U255, Université Pierre et Marie Curie, 20 Rue Leblanc 75908, Paris Cedex 15, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Rodrigues MM, Soares MBP, Vasconcelos JRC. Endogenous interleukin-4 downregulates the type 1 CD4 T cell-mediated immune response induced by intramuscular DNA immunization. J Interferon Cytokine Res 2002; 22:1137-41. [PMID: 12513913 DOI: 10.1089/10799900260442566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intramuscular (i.m.) administration of eukaryotic plasmid vectors containing foreign genes is a general immunization strategy capable of inducing protective type 1 immune responses against viral, bacterial, fungal, and parasitic infections. We have described that immunization with a plasmid containing a gene encoding a parasite antigen elicits specific type 1 protective immune responses against experimental infection with the human protozoan parasite Trypanosoma cruzi. However, we had evidence suggesting that DNA immunization concomitantly activated specific type 2 immune responses. To determine precisely the influence of the type 2 cytokine interleukin-4 (IL-4) during DNA immunization, we compared the immune responses of genetically modified IL-4-deficient or wild-type (wt) BALB/c mice. IL-4-deficient mice had a significantly lower ratio of specific serum IgG1/IgG2a, and on in vitro restimulation with antigen, their spleen cells secreted significantly higher amounts of interferon-gamma (IFN-gamma). In contrast, absence of IL-4 did not affect total serum antibody response, T cell proliferative responses, or activation of IFN-gamma-producing CD8(+) T cells. Our results suggested that in contrast to conventional adjuvants, such as alum and complete Freund's adjuvant, specific IgG1 in DNA-immunized BALB/c mice was highly dependent on IL-4. To our knowledge, our study provides the first evidence that endogenous IL-4 selectively downregulates the type 1 CD4(+) T cell-mediated immune response induced by i.m. genetic immunization, a fact that may have implications for the design of certain DNA vaccines.
Collapse
Affiliation(s)
- Mauricio M Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil, 04023-062.
| | | | | |
Collapse
|
48
|
Stetson DB, Mohrs M, Mallet-Designe V, Teyton L, Locksley RM. Rapid expansion and IL-4 expression by Leishmania-specific naive helper T cells in vivo. Immunity 2002; 17:191-200. [PMID: 12196290 DOI: 10.1016/s1074-7613(02)00363-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
CD4 T cells are pivotal for effective immunity, yet their initial differentiation into effector subsets after infection remains poorly defined. We examined CD4 T cells specific for the immunodominant Leishmania major LACK antigen using MHC/peptide tetramers and IL-4 reporter mice. Comprising approximately 15 cells/lymph node in naive mice, LACK-specific T cells expanded over 100-fold, and 70% acquired IL-4 expression by 96 hr. Despite their pathogenic role in susceptible mice, LACK-specific precursor frequency, expansion, and IL-4 expression were comparable between susceptible and resistant mice. When injected with unrelated antigen, Leishmania efficiently activated IL-4 expression from naive antigen-specific T cells. CD4 subset polarization in this highly characterized model occurs independently from IL-4 expression by naive T cells, which is activated indiscriminately after parasitism.
Collapse
Affiliation(s)
- Daniel B Stetson
- Howard Hughes Medical Institute, Department of Medicine, University of California, San Francisco, 94143, USA
| | | | | | | | | |
Collapse
|