1
|
Turrini R, Pabois A, Xenarios I, Coukos G, Delaloye JF, Doucey MA. TIE-2 expressing monocytes in human cancers. Oncoimmunology 2017; 6:e1303585. [PMID: 28507810 PMCID: PMC5414874 DOI: 10.1080/2162402x.2017.1303585] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAM) are well known as a key player in the tumor microenvironment, which support cancer progression. More recently, a lineage of monocytes characterized by the expression of the TIE-2/Tek angiopoietin receptor identified a subset of circulating and tumor-associated monocytes endowed with proangiogenic activity. TIE-2 expressing monocytes (TEM) were found both in humans and mice. Here, we review the phenotypes and functions of TEM reported so far in human cancer and their potential use as markers of cancer progression and metastasis. Finally, we discuss the therapeutic approaches currently used or proposed to target TEM.
Collapse
Affiliation(s)
- Riccardo Turrini
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Angélique Pabois
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ioannis Xenarios
- Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Marie-Agnès Doucey
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Ibberson M, Bron S, Guex N, Faes-van't Hull E, Ifticene-Treboux A, Henry L, Lehr HA, Delaloye JF, Coukos G, Xenarios I, Doucey MA. TIE-2 and VEGFR kinase activities drive immunosuppressive function of TIE-2-expressing monocytes in human breast tumors. Clin Cancer Res 2013; 19:3439-49. [PMID: 23649001 DOI: 10.1158/1078-0432.ccr-12-3181] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor-associated TIE-2-expressing monocytes (TEM) are highly proangiogenic cells critical for tumor vascularization. We previously showed that, in human breast cancer, TIE-2 and VEGFR pathways control proangiogenic activity of TEMs. Here, we examine the contribution of these pathways to immunosuppressive activity of TEMs. EXPERIMENTAL DESIGN We investigated the changes in immunosuppressive activity of TEMs and gene expression in response to specific kinase inhibitors of TIE-2 and VEGFR. The ability of tumor TEMs to suppress tumor-specific T-cell response mediated by tumor dendritic cells (DC) was measured in vitro. Characterization of TEM and DC phenotype in addition to their interaction with T cells was done using confocal microscopic images analysis of breast carcinomas. RESULTS TEMs from breast tumors are able to suppress tumor-specific immune responses. Importantly, proangiogenic and suppressive functions of TEMs are similarly driven by TIE-2 and VEGFR kinase activity. Furthermore, we show that tumor TEMs can function as antigen-presenting cells and elicit a weak proliferation of T cells. Blocking TIE-2 and VEGFR kinase activity induced TEMs to change their phenotype into cells with features of myeloid dendritic cells. We show that immunosuppressive activity of TEMs is associated with high CD86 surface expression and extensive engagement of T regulatory cells in breast tumors. TIE-2 and VEGFR kinase activity was also necessary to maintain high CD86 surface expression levels and to convert T cells into regulatory cells. CONCLUSIONS These results suggest that TEMs are plastic cells that can be reverted from suppressive, proangiogenic cells into cells that are able to mediate an antitumoral immune response.
Collapse
Affiliation(s)
- Mark Ibberson
- Vital-IT, Swiss Institute of Bioinformatics, CHUV, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Chen PW, Ksander BR. Termination of Systemic Immunity in the Presence of Intraocular Tumors: Influence of Ocular Immune Privilege on Tumor Vaccines. Curr Eye Res 2009; 31:43-55. [PMID: 16421019 DOI: 10.1080/02713680500477339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Ocular immune privilege preserves the visual axis by preventing the induction of sight-threatening nonspecific inflammation. Although privilege is essential for maintaining visual integrity, intraocular tumors exploit the privileged environment and grow progressively within the anterior chamber of the eye. Recently, a large number of laboratories have constructed genetically engineered tumor cell vaccines that express high levels of costimulatory signals. These vaccines are designed to bypass the normal pathways of T-cell activation and directly activate CD8+ tumor-specific T cells. In the following series of experiments, we determined whether a tumor cell vaccine that uses costimulatory signals (CD80 and IL-12) is capable of eliminating tumors within the immune-privileged anterior chamber. As expected, vaccine-immunized mice rejected subcutaneous flank tumors (a non-privileged site). However, the vaccine failed to protect mice from even a small number of tumor cells transplanted into the immune-privileged anterior chamber. Surprisingly, immunized mice that were simultaneously challenged with subcutaneous and anterior chamber tumors were unable to eliminate tumors at either site. The failure of systemic protective immunity coincided with the loss of tumor-specific delayed hypersensitivity and cytotoxic T cells. We conclude that tumor cell vaccines that induce complete protection against tumors in non-immune-privileged sites fail to protect against the same tumor within an ocular immune-privileged site. Moreover, a tumor that escapes elimination within the eye can terminate systemic protective immunity that is induced by the tumor cell vaccine.
Collapse
Affiliation(s)
- Peter W Chen
- Department of Ophthalmology, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9057, USA.
| | | |
Collapse
|
4
|
Abstract
SUMMARY Despite the expression of antigens by tumor cells, spontaneous immune-mediated rejection of cancer seems to be a rare event. T-cell receptor engagement by peptide/major histocompatibility complexes constitutes the main signal for the activation of naive T cells but is not sufficient to initiate a productive generation and maintenance of effector cells. Full activation of T cells requires additional signals driven by costimulatory molecules present on activated antigen-presenting cells but rarely on tumors. Following the discovery of B7-1 (CD80), several other costimulatory molecules have been shown to contribute to T-cell activation and have relevance for improving anti-tumor immunity. Moreover, increasing the understanding of coinhibitory receptors has highlighted key additional pathways that can dominantly inhibit anti-tumor T-cell function. Improving positive costimulation, and interfering with negative regulation, continues to represent an attractive immunotherapeutic approach for the treatment of cancer. This review focuses upon those pathways with the highest potential for clinical application in human cancer patients.
Collapse
Affiliation(s)
- Gregory Driessens
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Justin Kline
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | - Thomas F. Gajewski
- Department of Pathology, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
5
|
Driessen WHP, Ozawa MG, Arap W, Pasqualini R. Ligand-directed cancer gene therapy to angiogenic vasculature. ADVANCES IN GENETICS 2009; 67:103-121. [PMID: 19914451 PMCID: PMC7172741 DOI: 10.1016/s0065-2660(09)67004-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Gene therapy strategies in cancer have remained an active area of preclinical and clinical research. One of the current limitations to successful trials is the relative transduction efficiency to produce a therapeutic effect. While intratumoral injections are the mainstay of many treatment regimens to date, this approach is hindered by hydrostatic pressures within the tumor and is not always applicable to all tumor subtypes. Vascular-targeting strategies introduce an alternative method to deliver vectors with higher local concentrations and minimization of systemic toxicity. Moreover, therapeutic targeting of angiogenic vasculature often leads to enhanced bystander effects, improving efficacy. While identification of functional and systemically accessible molecular targets is challenging, approaches, such as in vivo phage display and phage-based viral delivery vectors, provide a platform upon which vascular targeting of vectors may become a viable and translational approach.
Collapse
Affiliation(s)
- Wouter H P Driessen
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Michael G Ozawa
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wadih Arap
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Renata Pasqualini
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
6
|
Rakhmilevich AL, Buhtoiarov IN, Malkovsky M, Sondel PM. CD40 ligation in vivo can induce T cell independent antitumor effects even against immunogenic tumors. Cancer Immunol Immunother 2008; 57:1151-60. [PMID: 18214476 PMCID: PMC11031017 DOI: 10.1007/s00262-007-0447-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Accepted: 12/20/2007] [Indexed: 11/25/2022]
Abstract
Antitumor effects of CD40 ligation appear to involve distinct antitumor effector cells in different experimental models. In this study, we tested whether T cells were required for antitumor effects of agonistic anti-CD40 mAb (alphaCD40) against immunogenic versus poorly immunogenic tumors. Treatment of mice bearing poorly immunogenic B16 melanoma and its more immunogenic variant, B16-hsp72.1, with alphaCD40 resulted in a similar level of tumor growth suppression. Depletion of T cells did not reduce the antitumor effects in these 2 tumor models. To generate antitumor T cell responses, C57BL/6 mice were immunized with irradiated B16-hsp72.1. Treatment of these vaccinated mice challenged with a high dose of B16-hsp72.1 tumor cells with alphaCD40 induced tumor growth suppression, which was reduced by T-cell depletion, demonstrating that T cells were involved in the antitumor effect of alphaCD40. However, immunized mice depleted of T cells and treated with alphaCD40 were still able to suppress tumor growth as compared to tumor growth in immunized, T cell-depleted mice not treated with alphaCD40, suggesting that T cells were not required for the antitumor effect of alphaCD40. To confirm a lack of correlation between tumor immunogenicity and T-cell requirement in antitumor effects of CD40 ligation, we found that alphaCD40 induced tumor growth suppression in nude and SCID/beige mice bearing highly immunogenic tumors such as Meth A sarcoma, suggesting that macrophages may play a role. Indeed, both poorly immunogenic and highly immunogenic tumors were sensitive to in vitro growth inhibition by macrophages from alphaCD40-treated mice. Taken together, our results indicate that antitumor effects induced by alphaCD40, even against immunogenic tumors, can be observed in the absence of T cells and may involve macrophages.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- CD40 Antigens/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Cancer Vaccines/therapeutic use
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/therapy
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease Models, Animal
- Female
- Immunotherapy
- Kidney Neoplasms/immunology
- Kidney Neoplasms/therapy
- Leukemia L5178/immunology
- Leukemia L5178/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Nude
- Mice, SCID
- Neoplasm Transplantation/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Alexander L Rakhmilevich
- The Department of Human Oncology, University of Wisconsin, K4/413 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792, USA.
| | | | | | | |
Collapse
|
7
|
Yang ZM, Li EM, Lai BC, Wang YL, Si LS. Anti-CD3 scFv-B7.1 fusion protein expressed on the surface of HeLa cells provokes potent T-lymphocyte activation and cytotoxicityThis paper is one of a selection of papers in this Special Issue, entitled International Symposium on Recent Advances in Molecular, Clinical, and Social Medicine, and has undergone the Journal's usual peer-review process. Biochem Cell Biol 2007; 85:196-202. [PMID: 17534400 DOI: 10.1139/o07-013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The targeting of tumor cells by cytotoxic T lymphocytes is a promising strategy for biotherapy, but T cells require 2 signals via the T-cell receptor – CD3 complex and CD28 molecules for activation. To bridge the gap between cytotoxic T lymphocytes and tumor cells, our objective in this study was to describe the construction and the cell surface-anchored expression of a fusion protein, anti-CD3 scFv-B7.1, derived from inserting a fusion gene encoding anti-CD3 scFv and the extra-cellular domain of B7.1 fused by the splicing by overlap extension method into a mammalian expression vector, pDisplay. Transfection of the recombinant vector by electroporation into HeLa cells resulted in the production of protein migrating at approximately 57 kDa under reducing conditions. The expressed fusion protein could bind to T lymphocytes and induce strong T-cell activation. Meanwhile, a potent cytotoxicity was induced in the mixed culture of T-cell-modified tumor cells in a 96 h methyl-thiazolyl-diphenyl tetrazolium bromide assay. Our results indicate that this bifunctional protein, through activating T lymphocytes to lyse homologous human carcinomas, may be of potential value for T-cell-based immunotherapeutical treatment protocols in vivo.
Collapse
Affiliation(s)
- Zhang-Min Yang
- Department of Biochemistry and Molecular Biology, Medical College, Shantou University, Shantou 515041, PR China.
| | | | | | | | | |
Collapse
|
8
|
Jabrane-Ferrat N, Campbell MJ, Esserman LJ, Peterlin BM. Challenge with mammary tumor cells expressing MHC class II and CD80 prevents the development of spontaneously arising tumors in MMTV-neu transgenic mice. Cancer Gene Ther 2006; 13:1002-10. [PMID: 16841083 DOI: 10.1038/sj.cgt.7700974] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The HER-2/Neu oncogene has been implicated in human and mouse breast cancer. Indeed, transgenic MMTV-neu mice expressing this oncogene from the mammary tumor virus long terminal repeat develop spontaneous mammary tumors and die within 1 year of life. We have expressed the class II transactivator (CIITA) and/or the costimulatory molecule CD80 (B7.1) in a mammary carcinoma cell line (MCNeuA) derived from these mice. Class II transactivator directs the expression of MHC class II and the machinery for antigen processing and presentation by this pathway. When injected into MMTV-neu mice, tumor cells expressing CD80 or CD80 and CIITA, were rejected completely. In addition, following the rejection of dual expressing cells, 75% of the mice were protected against the development of subsequent spontaneous tumors. Cells expressing only CD80 or CIITA were not as effective as antitumor vaccines in preventing the development of spontaneous tumors. Thus, converting cancer cells into antigen presenting cells could represent an effective immunotherapy for breast cancer.
Collapse
MESH Headings
- Animals
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- B7-1 Antigen/metabolism
- Blotting, Western
- Cell Line
- Cell Line, Tumor
- Female
- Flow Cytometry
- Genes, MHC Class II/genetics
- Genes, MHC Class II/immunology
- Humans
- Immunohistochemistry
- Immunoprecipitation
- Lymphocytes/immunology
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Transgenic
- Nuclear Proteins/genetics
- Nuclear Proteins/immunology
- Nuclear Proteins/metabolism
- Rats
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Trans-Activators/genetics
- Trans-Activators/immunology
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- N Jabrane-Ferrat
- Institut de Sciences et Technologies du Medicament de Toulouse, CNRS-Pierre Fabre, Toulouse, France
| | | | | | | |
Collapse
|
9
|
Kudo-Saito C, Hodge JW, Kwak H, Kim-Schulze S, Schlom J, Kaufman HL. 4-1BB ligand enhances tumor-specific immunity of poxvirus vaccines. Vaccine 2006; 24:4975-86. [PMID: 16621183 PMCID: PMC1865095 DOI: 10.1016/j.vaccine.2006.03.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 03/14/2006] [Accepted: 03/16/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE Recombinant poxvirus vaccines have been explored as tumor vaccines. The immunogenicity of these vaccines can be enhanced by co-expressing costimulatory molecules and tumor-associated antigens. While the B7-CD28 interaction has been most comprehensively investigated, other costimulatory molecules utilize different signaling pathways and might provide further cooperation in T cell priming and survival. 4-1BB (CD137) is a TNF family member and is critical for activation and long-term maintenance of primed T cells. This study was conducted to determine if a poxvirus expressing the ligand for 4-1BB (4-1BBL) could further improve the immune and therapeutic responses of a previously reported poxvirus vaccine expressing a triad of costimulatory molecules (B7.1, ICAM-1, and LFA-3). EXPERIMENTAL DESIGN A recombinant vaccinia virus expressing 4-1BBL was generated and characterized in an in vitro infection system. This vaccine was then used alone or in combination with a vaccinia virus expressing CEA, B7.1, ICAM-1, and LFA-3 in CEA-transgenic mice bearing established MC38 tumors. Tumor growth and immune responses against CEA and other tumor-associated antigens were determined. The level of anti-apoptotic proteins in responding T cells was determined by flow cytometry on tetramer selected T cells. RESULTS The combination of 4-1BBL with B7.1-based poxvirus vaccination resulted in significantly enhanced therapeutic effects against CEA-expressing tumors in a CEA-transgenic mouse model. This was associated with an increased level of CEA-specific CD4(+) and CD8(+) T cell responses, induction of antigen spreading to p53 and gp70, increased accumulation of CEA-specific T cells in the tumor microenvironment, and increased expression of bcl-X(L) and bcl-2 in CD4(+) and CD8(+) T cells in vaccinated mice. CONCLUSION 4-1BBL cooperates with B7 in enhancing anti-tumor and immunologic responses in a recombinant poxvirus vaccine model. The inclusion of costimulatory molecules targeting distinct T cell signaling pathways provides a mechanism for enhancing the therapeutic effectiveness of tumor vaccines.
Collapse
Affiliation(s)
- Chie Kudo-Saito
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| | - Heesun Kwak
- The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032
| | - Seunghee Kim-Schulze
- The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| | - Howard L. Kaufman
- The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032
- To whom correspondence should be addressed, at The Tumor Immunology Laboratory, Columbia University, 177 Fort Washington Avenue, New York, NY 10032; E-mail:
| |
Collapse
|
10
|
Triozzi PL, Allen KO, Carlisle RR, Craig M, LoBuglio AF, Conry RM. Phase I study of the intratumoral administration of recombinant canarypox viruses expressing B7.1 and interleukin 12 in patients with metastatic melanoma. Clin Cancer Res 2005; 11:4168-75. [PMID: 15930353 DOI: 10.1158/1078-0432.ccr-04-2283] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The objective of this study was to evaluate the safety and activity of the intratumoral administration of the immune costimulatory molecule, B7.1, encoded by a vector derived from the canarypox virus, ALVAC (ALVAC-B7.1), alone and with the intratumoral injection of ALVAC encoding the immune-stimulatory cytokine, interleukin 12 (ALVAC-IL-12). Fourteen patients with metastatic melanoma who had s.c. nodules received intratumoral injections on days 1, 4, 8, and 11. Nine patients were given escalating doses of up to 25 x 10(8) plaque-forming units of ALVAC-B7.1. Five patients were given 25 x 10(8) plaque-forming units of ALVAC-B7.1 combined with ALVAC-IL-12 50% tissue culture infective dose of 2 x 10(6). Toxicity was mild to moderate and consisted of inflammatory reactions at the injection site and fever, chills, myalgia, and fatigue. Higher levels of B7.1 mRNA were observed in ALVAC-B7.1-injected tumors compared with saline-injected control tumors. Higher levels of intratumoral vascular endothelial growth factor and IL-10, cytokines with immune suppressive activities, were also observed in ALVAC-B7.1- and ALVAC-IL-12-injected tumors compared with saline-injected controls. Serum levels of vascular endothelial growth factor increased at day 18 and returned to baseline at day 43. All patients developed antibody to ALVAC. Intratumoral IL-12 and IFN-gamma mRNA decreased. Changes in serum IL-12 and IFN-gamma levels were not observed. Tumor regressions were not observed. The intratumoral injections of ALVAC-B7.1 and ALVAC-IL-12 were well tolerated at these dose levels and at this schedule and resulted in measurable biological response. This response included the production of factors that may suppress the antitumor immunologic activity of these vectors.
Collapse
Affiliation(s)
- Pierre L Triozzi
- The University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, Alabama 35294-3300, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Esplugues E, Vega-Ramos J, Cartoixà D, Vazquez BN, Salaet I, Engel P, Lauzurica P. Induction of tumor NK-cell immunity by anti-CD69 antibody therapy. Blood 2005; 105:4399-406. [PMID: 15692061 DOI: 10.1182/blood-2004-10-3854] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Abstract
The leukocyte activation marker CD69 is a novel regulator of the immune response, modulating the production of cytokines including transforming growth factor-β (TGF-β). We have generated an antimurine CD69 monoclonal antibody (mAb), CD69.2.2, which down-regulates CD69 expression in vivo but does not deplete CD69-expressing cells. Therapeutic administration of CD69.2.2 to wild-type mice induces significant natural killer (NK) cell–dependent antitumor responses to major histocompatibility complex (MHC) class I low RMA-S lymphomas and to RM-1 prostatic carcinoma lung metastases. These in vivo antitumor responses are comparable to those seen in CD69-/- mice. Enhanced host NK cytotoxic activity correlates with a reduction in NK-cell TGF-β production and is independent of tumor priming. In vitro studies demonstrate the novel ability of anti-CD69 mAbs to activate resting NK cells in an Fc receptor–independent manner, resulting in a substantial increase in both NK-cell cytolytic activity and interferon γ (IFNγ) production. Modulation of the innate immune system with monoclonal antibodies to host CD69 thus provides a novel means to antagonize tumor growth and metastasis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Cytotoxicity, Immunologic
- Immunity/drug effects
- Immunotherapy/methods
- Interferon-gamma/biosynthesis
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lectins, C-Type
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lymphocyte Activation/drug effects
- Lymphoma/drug therapy
- Lymphoma/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Neoplasms/immunology
- Neoplasms/therapy
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/pathology
Collapse
Affiliation(s)
- Enric Esplugues
- Departamento de Fisiología, Universidad de Barcelona, Av Diagonal, 645 Barcelona 08028, Spain
| | | | | | | | | | | | | |
Collapse
|
12
|
Shi G, Mao J, Yu G, Zhang J, Wu J. Tumor vaccine based on cell surface expression of DcR3/TR6. THE JOURNAL OF IMMUNOLOGY 2005; 174:4727-35. [PMID: 15814697 DOI: 10.4049/jimmunol.174.8.4727] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
DcR3/TR6, a secreted protein belonging to the TNF receptor superfamily, interacts with lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entrance mediator (LIGHT), Fas ligand (FasL), and TL1A, all members of the TNF superfamily. Solid-phase TR6 can trigger reverse signaling of LIGHT and FasL expressed on T cells, and lead to T cell costimulation. In this study, we engineered tumor cells to express cell surface TR6 and used these cells as a tumor vaccine. We demonstrated that mastocytoma P815 cells expressing surface TR6 (TR6-P815) effectively augmented the T cells response in vitro and ex vivo in terms of proliferation, as well as IL-2 and IFN-gamma secretion. TR6-P815 cells had reduced tumorigenicity compared with parental P815 cells. When inactivated TR6-P815 cells were employed as a vaccine, they protected the mice from challenge with live parental P815 cells, and eliminated established P815 tumors. The cell surface TR6-based tumor vaccine was also effective against low antigenicity tumors, such as B16 melanoma; co-administration of bacillus Calmette-Guérin further enhanced the vaccine's efficacy. Thus, cell surface TR6 expression is a useful addition to our tumor vaccine arsenal.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/isolation & purification
- Cancer Vaccines/pharmacology
- Cell Line, Tumor
- Cell Membrane/immunology
- Female
- Humans
- Immunologic Factors/administration & dosage
- In Vitro Techniques
- Lymphocyte Activation
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Mice, Nude
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor, Member 6b
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Guixiu Shi
- Laboratory of Immunology, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
13
|
Tarazona R, Casado JG, Soto R, DelaRosa O, Peralbo E, Rioja L, Peña J, Solana R. Expression of NK-associated receptors on cytotoxic T cells from melanoma patients: a two-edged sword? Cancer Immunol Immunother 2004; 53:911-24. [PMID: 15127235 PMCID: PMC11032847 DOI: 10.1007/s00262-004-0507-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Accepted: 01/27/2004] [Indexed: 11/29/2022]
Abstract
The coexistence of tumor progression with a tumor-specific immune response constitutes a major paradox of tumor immunity. During the last decade, the presence of cytotoxic T lymphocytes (CTLs) recognising melanoma-associated antigens has been unequivocally demonstrated in numerous different in vivo and in vitro models. However, most often these melanoma-specific T lymphocytes do not control tumor growth. Several mechanisms that involve changes in melanoma phenotype and/or in T-cell differentiation and function could explain the inability of the immune response to control melanoma. In the last few years it has been demonstrated that cellular cytotoxicity is the result of a balance between activating signals triggered by the TCR and costimulatory molecules and inhibitory signals triggered by inhibitory receptors expressed by the CTL. Because the final outcome of the immune response against melanoma depends on the balance between activating and inhibitory signals, the expression de novo on melanoma cells of ligands for inhibitory NKRs and the down-regulation of costimulatory molecules may favor the escape of tumor cells from immunosurveillance. In this paper we review how altered expression of molecules required for T-cell costimulation could result in impaired lysis of melanoma. The modulation of antimelanoma T-cell responses by a group of receptors originally described on NK cells (NK-associated receptors) but which are now known also to be expressed on a subset of cytolytic effector cells is reviewed. We hypothesize that the expression of ligands for NKRs on melanoma cells may contribute to T-cell-mediated immune responses against melanoma either enhancing or inhibiting activation and differentiation to effector cells. Blocking inhibitory receptors or increasing activating receptors could result in new strategies to improve T-cell-mediated rejection of melanoma.
Collapse
Affiliation(s)
- Raquel Tarazona
- Department of Physiology, Immunology Unit, Veterinary Faculty, University of Extremadura, Cáceres, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Pawelec G. Immunotherapy and immunoselection -- tumour escape as the final hurdle. FEBS Lett 2004; 567:63-6. [PMID: 15165894 DOI: 10.1016/j.febslet.2004.02.091] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2004] [Accepted: 02/11/2004] [Indexed: 11/16/2022]
Abstract
Tumours are immunogenic and are commonly infiltrated by anti-cancer effector cells. Why, then, are they not completely rejected by the host? Unfortunately, tumours are Darwinian paragons, winning the battle against the forces of natural immune selection. Some of the latter can even act as double-edged swords, actually being subverted to become pro-tumorigenic. Prevention or reversal of tumour escape from the immune response therefore offers the possibility of reconstituting effective anti-tumour immunity and remains the major challenge for 21st century tumour immunology.
Collapse
Affiliation(s)
- Graham Pawelec
- Section for Transplantation Immunology and Immunohaematology, Center for Medical Research, University of Tübingen Medical School, ZMF, Waldhörnlestr. 22, D-72072 Tübingen, Germany.
| |
Collapse
|