151
|
Oxidative stress, polarization of macrophages and tumour angiogenesis: Efficacy of caffeic acid. Chem Biol Interact 2016; 256:111-24. [PMID: 27378625 DOI: 10.1016/j.cbi.2016.06.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/23/2016] [Accepted: 06/29/2016] [Indexed: 12/22/2022]
Abstract
Macrophage polarization is a process when macrophage expresses different functional programs in response to microenvironmental signals and two extreme forms exist; M1 and M2 macrophages. M1 macrophages are highly microbicidal and anticancer with enhanced ability to kill and phagocytose pathogens, upregulate pro-inflammatory cytokines and reactive molecular species, and present antigens; M2 macrophages and the related tumour associated macrophages (TAMs) regulate tissue remodelling and promote tissue repair and angiogenesis and can amplification of metabolic pathways that can suppress adaptive immune responses. It is demonstrated that ROS production, critical for the activation and functions of M1 macrophages, is necessary for the differentiation of M2 macrophages and TAMs, and that antioxidant therapy blocks TAMs differentiation and tumorigenesis in mouse models of cancer. In order to study how caffeic acid (CA), a natural antioxidant, affects macrophage function, polarization, angiogenesis and tumour growth we injected mice with Ehrlich ascites tumour (EAT) cells and treated them for 10 days with CA in a dose of 40 and/or 80 mg kg(-1.) Macrophage polarization was further characterized by quantifying secreted pro- and anti-inflammatory cytokines, nitric oxide and arginase 1 activity. CA may increase the cytotoxic actions of M1 macrophages and inhibit tumour growth; inhibitory activity on TAMs may be mediated through its antioxidative activity. Taken together, we conclude that the antitumour activity of CA was the result of the synergistic activities of different mechanisms by which CA acts on proliferation, angiogenesis, immunomodulation and survival. The continuous administration of CA efficiently blocked the occurrence of TAMs and markedly suppressed tumorigenesis in mouse cancer models. Targeting TAMs by antioxidants can be a potentially effective method for cancer treatment.
Collapse
|
152
|
Ding ZC, Liu C, Cao Y, Habtetsion T, Kuczma M, Pi W, Kong H, Cacan E, Greer SF, Cui Y, Blazar BR, Munn DH, Zhou G. IL-7 signaling imparts polyfunctionality and stemness potential to CD4(+) T cells. Oncoimmunology 2016; 5:e1171445. [PMID: 27471650 DOI: 10.1080/2162402x.2016.1171445] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 12/21/2022] Open
Abstract
The functional status of CD4(+) T cells is a critical determinant of antitumor immunity. Polyfunctional CD4(+) T cells possess the ability to concomitantly produce multiple Th1-type cytokines, exhibiting a functional attribute desirable for cancer immunotherapy. However, the mechanisms by which these cells are induced are neither defined nor it is clear if these cells can be used therapeutically to treat cancer. Here, we report that CD4(+) T cells exposed to exogenous IL-7 during antigenic stimulation can acquire a polyfunctional phenotype, characterized by their ability to simultaneously express IFNγ, IL-2, TNFα and granzyme B. This IL-7-driven polyfunctional phenotype was associated with increased histone acetylation in the promoters of the effector genes, indicative of increased chromatin accessibility. Moreover, forced expression of a constitutively active (CA) form of STAT5 recapitulated IL-7 in inducing CD4(+) T-cell polyfunctionality. Conversely, the expression of a dominant negative (DN) form of STAT5 abolished the ability of IL-7 to induce polyfunctional CD4(+) T cells. These in-vitro-generated polyfunctional CD4(+) T cells can traffic to tumor and expand intratumorally in response to immunization. Importantly, adoptive transfer of polyfunctional CD4(+) T cells following lymphodepletive chemotherapy was able to eradicate large established tumors. This beneficial outcome was associated with the occurrence of antigen epitope spreading, activation of the endogenous CD8(+) T cells and persistence of donor CD4(+) T cells exhibiting memory stem cell attributes. These findings indicate that IL-7 signaling can impart polyfunctionality and stemness potential to CD4(+) T cells, revealing a previously unknown property of IL-7 that can be exploited in adoptive T-cell immunotherapy.
Collapse
Affiliation(s)
- Zhi-Chun Ding
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University , Augusta, GA, USA
| | - Chufeng Liu
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University, Augusta, GA, USA; Department of Orthodontics, Guangdong Provincial Stomatological Hospital, Southern Medical University, Guangzhou, PR, China
| | - Yang Cao
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University, Augusta, GA, USA; Department of Orthodontics, Guangdong Provincial Key Laboratory of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, PR, China
| | - Tsadik Habtetsion
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University , Augusta, GA, USA
| | - Michal Kuczma
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University , Augusta, GA, USA
| | - Wenhu Pi
- Department of Biochemistry and Molecular Biology, Augusta University , Augusta, GA, USA
| | - Heng Kong
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University, Augusta, GA, USA; Department of Thyroid and Breast Surgery, Shenzhen Nanshan District People's Hospital, Shenzhen, Guangzhou, PR, China
| | - Ercan Cacan
- Department of Biology, Georgia State University , Atlanta, GA, USA
| | - Susanna F Greer
- Department of Biology, Georgia State University , Atlanta, GA, USA
| | - Yan Cui
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University , Augusta, GA, USA
| | - Bruce R Blazar
- Department of Pediatrics and Division of Blood and Marrow Transplantation, University of Minnesota , Minneapolis, MN, USA
| | - David H Munn
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University , Augusta, GA, USA
| | - Gang Zhou
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Cancer Center, Augusta University , Augusta, GA, USA
| |
Collapse
|
153
|
Nabizadeh JA, Manthey HD, Steyn FJ, Chen W, Widiapradja A, Md Akhir FN, Boyle GM, Taylor SM, Woodruff TM, Rolfe BE. The Complement C3a Receptor Contributes to Melanoma Tumorigenesis by Inhibiting Neutrophil and CD4+ T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2016; 196:4783-92. [DOI: 10.4049/jimmunol.1600210] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/21/2016] [Indexed: 01/28/2023]
|
154
|
Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, Gouw AM, Baylot V, Gütgemann I, Eilers M, Felsher DW. MYC regulates the antitumor immune response through CD47 and PD-L1. Science 2016; 352:227-31. [PMID: 26966191 DOI: 10.1126/science.aac9935] [Citation(s) in RCA: 1028] [Impact Index Per Article: 114.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 02/18/2016] [Indexed: 11/03/2022]
Abstract
The MYC oncogene codes for a transcription factor that is overexpressed in many human cancers. Here we show that MYC regulates the expression of two immune checkpoint proteins on the tumor cell surface: the innate immune regulator CD47 (cluster of differentiation 47) and the adaptive immune checkpoint PD-L1 (programmed death-ligand 1). Suppression of MYC in mouse tumors and human tumor cells caused a reduction in the levels of CD47 and PD-L1 messenger RNA and protein. MYC was found to bind directly to the promoters of the Cd47 and Pd-l1 genes. MYC inactivation in mouse tumors down-regulated CD47 and PD-L1 expression and enhanced the antitumor immune response. In contrast, when MYC was inactivated in tumors with enforced expression of CD47 or PD-L1, the immune response was suppressed, and tumors continued to grow. Thus, MYC appears to initiate and maintain tumorigenesis, in part, through the modulation of immune regulatory molecules.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ling Tong
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yulin Li
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel Do
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Susanne Walz
- Comprehensive Cancer Center Mainfranken, Core Unit Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Kelly N Fitzgerald
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arvin M Gouw
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Virginie Baylot
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ines Gütgemann
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA. Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Martin Eilers
- Comprehensive Cancer Center Mainfranken, Core Unit Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany. Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
155
|
Yao X, Dong Z, Zhang Q, Wang Q, Lai D. Epithelial ovarian cancer stem-like cells expressing α-gal epitopes increase the immunogenicity of tumor associated antigens. BMC Cancer 2015; 15:956. [PMID: 26673159 PMCID: PMC4682262 DOI: 10.1186/s12885-015-1973-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/04/2015] [Indexed: 12/18/2022] Open
Abstract
Background As ovarian cancer stem cells (CSCs) are responsible for tumor initiation, invasion, metastasis, and chemo-resistance, new stratagems that selectively target ovarian CSCs are critically significant. Our previous work have demonstrated that ovarian cancer spheroid cells are tumorigenic and chemo-resistant, and have the properties of ovarian CSCs. Herein, we hypothesized that expressing α-gal epitopes on ovarian spheroid cells may help eliminate CSCs and improve the outcome of therapeutic intervention for ovarian cancer patients. Methods Lentivirus-mediated transfer of a pig α(1,3)galactosyltransferase [α1,3GT] enzyme gene into human ovarian cell line SKOV3 cells formed α-gal epitope-expressing cells (SKOV3-gal cells), and then these cells were maintained in a serum-free culture system to form SKOV3-gal spheroid cells. Efficacy of this cell vaccine was demonstrated in α1,3GT knockout mice (α1,3GT KO mice). Results The antibody titers to α-gal epitopes measured by ELISA were significantly increased in α1,3GT KO mice after immunization with SKOV3-gal spheroid cells. Furthermore, compared with the non-immunized KO mice, the SKOV3 tumors grafted under renal capsules of KO mice immunized with SKOV3-gal spheroid cells grew slower and began to shrink on day 12. Western blot analysis also showed that immunized KO mice can produce effective antibody against certain tumor associated antigens (TAAs) derived from both SKOV3 cells and SKOV3 spheroid cells. The TAAs were further investigated by mass spectrometry and RNA interference (RNAi) technology. The results suggested that antibodies responding to protein c-erbB-2 may be raised in the sera of the mice after immunization with SKOV3-gal spheroid cells. Ultimately, vaccination with SKOV3-gal spheroid cells induced more CD3 + CD4 + T cells in the spleen of immunized mice than non-immunized KO mice. Conclusions The results suggest that vaccination using ovarian cancer stem-like cells engineered to express α-gal epitopes may be a novel strategy for treatment of ovarian cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1973-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaofen Yao
- The Center of Research Laboratory, and Department of Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Zhangli Dong
- The Center of Research Laboratory, and Department of Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Qiuwan Zhang
- The Center of Research Laboratory, and Department of Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Qian Wang
- The Center of Research Laboratory, and Department of Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Dongmei Lai
- The Center of Research Laboratory, and Department of Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
156
|
Chai SJ, Yap YY, Foo YC, Yap LF, Ponniah S, Teo SH, Cheong SC, Patel V, Lim KP. Identification of Four-Jointed Box 1 (FJX1)-Specific Peptides for Immunotherapy of Nasopharyngeal Carcinoma. PLoS One 2015; 10:e0130464. [PMID: 26536470 PMCID: PMC4633155 DOI: 10.1371/journal.pone.0130464] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/20/2015] [Indexed: 01/02/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is highly prevalent in South East Asia and China. The poor outcome is due to late presentation, recurrence, distant metastasis and limited therapeutic options. For improved treatment outcome, immunotherapeutic approaches focusing on dendritic and autologous cytotoxic T-cell based therapies have been developed, but cost and infrastructure remain barriers for implementing these in low-resource settings. As our prior observations had found that four-jointed box 1 (FJX1), a tumor antigen, is overexpressed in NPCs, we investigated if short 9-20 amino acid sequence specific peptides matching to FJX1 requiring only intramuscular immunization to train host immune systems would be a better treatment option for this disease. Thus, we designed 8 FJX1-specific peptides and implemented an assay system to first, assess the binding of these peptides to HLA-A2 molecules on T2 cells. After, ELISPOT assays were used to determine the peptides immunogenicity and ability to induce potential cytotoxicity activity towards cancer cells. Also, T-cell proliferation assay was used to evaluate the potential of MHC class II peptides to stimulate the expansion of isolated T-cells. Our results demonstrate that these peptides are immunogenic and peptide stimulated T-cells were able to induce peptide-specific cytolytic activity specifically against FJX1-expressing cancer cells. In addition, we demonstrated that the MHC class II peptides were capable of inducing T-cell proliferation. Our results suggest that these peptides are capable of inducing specific cytotoxic cytokines secretion against FJX1-expressing cancer cells and serve as a potential vaccine-based therapy for NPC patients.
Collapse
Affiliation(s)
- San Jiun Chai
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Yoke Yeow Yap
- Department of Surgery, Clinical Campus Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Yoke Ching Foo
- Department of Oncology, Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Lee Fah Yap
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Sathibalan Ponniah
- Department of Surgery, Cancer Vaccine Development Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Soo Hwang Teo
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Sok Ching Cheong
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Vyomesh Patel
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
| | - Kue Peng Lim
- Cancer Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang Jaya, Selangor, Malaysia
- * E-mail:
| |
Collapse
|
157
|
Haabeth OA, Tveita A, Fauskanger M, Hennig K, Hofgaard PO, Bogen B. Idiotype-specific CD4(+) T cells eradicate disseminated myeloma. Leukemia 2015; 30:1216-20. [PMID: 26449664 DOI: 10.1038/leu.2015.278] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- O A Haabeth
- Centre for Immune Regulation, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - A Tveita
- Centre for Immune Regulation, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - M Fauskanger
- Centre for Immune Regulation, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - K Hennig
- Centre for Immune Regulation, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - P O Hofgaard
- KG Jebsen Centre for Influenza Vaccine Research, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - B Bogen
- Centre for Immune Regulation, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway.,KG Jebsen Centre for Influenza Vaccine Research, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
158
|
Turner N, Tran B, Tran PV, Sinnathamby M, Wong HL, Jones I, Croxford M, Desai J, Tie J, Field KM, Kosmider S, Bae S, Gibbs P. Primary Tumor Resection in Patients With Metastatic Colorectal Cancer Is Associated With Reversal of Systemic Inflammation and Improved Survival. Clin Colorectal Cancer 2015; 14:185-91. [DOI: 10.1016/j.clcc.2015.02.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/17/2015] [Accepted: 02/26/2015] [Indexed: 12/31/2022]
|
159
|
Binsfeld M, Hannon M, Otjacques E, Humblet-Baron S, Baudoux E, Beguin Y, Baron F, Caers J. Impact of the immunomodulating peptide thymosin alpha 1 on multiple myeloma and immune recovery after hematopoietic stem cell transplantation. Cancer Immunol Immunother 2015; 64:989-98. [PMID: 25971542 PMCID: PMC11028767 DOI: 10.1007/s00262-015-1708-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/04/2015] [Indexed: 10/23/2022]
Abstract
Multiple myeloma (MM) is characterized by the accumulation of monoclonal plasma cells in the bone marrow and causes several immune alterations in patients. Thymosin α1 (Tα1) is a thymic peptide that has been associated with immuno-stimulating properties. In addition, this peptide exerts anti-tumor effects in several cancer types. Beneficial effects of Tα1 administration have also been shown on immune reconstitution after hematopoietic stem cell transplantation (HSCT), a current treatment modality in hematological malignancies including MM. In this study, we observed a slight reduction in the proliferation of murine and human MM cell lines in the presence of Tα1 in vitro. However, using two immunocompetent murine MM models (5TGM1 and MOPC315.BM), we did not observe any impact of Tα1 administration on MM development in vivo. Furthermore, no beneficial effects of Tα1 treatment were observed on lymphocyte immune reconstitution after transfusion of human hematopoietic stem cells into immunodeficient mice. In conclusion, despite direct effects of Tα1 on human MM cell line proliferation in vitro, Tα1 did not exert anti-myeloma effects in vivo in the two murine models tested. Moreover, Tα1 failed to improve immune recovery in a xenogeneic HSCT model.
Collapse
Affiliation(s)
- Marilène Binsfeld
- Laboratory of Hematology, GIGA-Research, University of Liège, Bat. B34, CHU of Liège, Avenue de l'Hôpital, 1, 4000, Liège, Belgium,
| | | | | | | | | | | | | | | |
Collapse
|
160
|
Haabeth OAW, Lorvik KB, Yagita H, Bogen B, Corthay A. Interleukin-1 is required for cancer eradication mediated by tumor-specific Th1 cells. Oncoimmunology 2015; 5:e1039763. [PMID: 26942052 PMCID: PMC4760324 DOI: 10.1080/2162402x.2015.1039763] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/01/2015] [Accepted: 04/06/2015] [Indexed: 12/21/2022] Open
Abstract
The role of inflammation in cancer is controversial as both tumor-promoting and tumor-suppressive aspects of inflammation have been reported. In particular, it has been shown that pro-inflammatory cytokines, like interleukin-1α (IL-1α), IL-1β, IL-6, and tumor necrosis factor α (TNFα), may either promote or suppress cancer. However, the cellular and molecular basis underlying these opposing outcomes remains enigmatic. Using mouse models for myeloma and lymphoma, we have recently reported that inflammation driven by tumor-specific T helper 1 (Th1) cells conferred protection against B-cell cancer and that interferon-γ (IFN-γ) was essential for this process. Here, we have investigated the contribution of several inflammatory mediators. Myeloma eradication by Th1 cells was not affected by inhibition of TNF-α, TNF-related weak inducer of apoptosis (TWEAK), or TNF-related apoptosis-inducing ligand (TRAIL). In contrast, cancer elimination by tumor-specific Th1 cells was severely impaired by the in vivo neutralization of both IL-1α and IL-1β (collectively named IL-1) with IL-1 receptor antagonist (IL-1Ra). The antitumor functions of tumor-specific Th1 cells and tumor-infiltrating macrophages were both affected by IL-1 neutralization. Secretion of the Th1-derived cytokines IL-2 and IFN-γ at the incipient tumor site was severely reduced by IL-1 blockade. Moreover, IL-1 was shown to synergize with IFN-γ for induction of tumoricidal activity in tumor-infiltrating macrophages. This synergy between IL-1 and IFN-γ may explain how inflammation, when driven by tumor-specific Th1 cells, represses rather than promotes cancer. Collectively, the data reveal a central role of inflammation, and more specifically of the canonical pro-inflammatory cytokine IL-1, in enhancing Th1-mediated immunity against cancer.
Collapse
Affiliation(s)
- Ole Audun Werner Haabeth
- Centre for Immune Regulation; University of Oslo and Oslo University Hospital Rikshospitalet ; Oslo, Norway
| | - Kristina Berg Lorvik
- Centre for Immune Regulation; University of Oslo and Oslo University Hospital Rikshospitalet ; Oslo, Norway
| | - Hideo Yagita
- Department of Immunology; Juntendo University School of Medicine ; Tokyo, Japan
| | - Bjarne Bogen
- Centre for Immune Regulation; University of Oslo and Oslo University Hospital Rikshospitalet; Oslo, Norway; K.G. Jebsen Centre for Influenza Vaccine Research; University of Oslo; Oslo, Norway
| | - Alexandre Corthay
- Centre for Immune Regulation; University of Oslo and Oslo University Hospital Rikshospitalet ; Oslo, Norway
| |
Collapse
|
161
|
Abstract
Research over the past decade has revealed the increasingly complex biologic features of the CD4(+) T-cell lineage. This T-cell subset, which was originally defined on the basis of helper activity in antibody responses, expresses receptors that recognize peptides that have been processed and presented by specialized antigen-presenting cells. At the core of the adaptive immune response, CD4 T cells display a large degree of plasticity and the ability to differentiate into multiple sublineages in response to developmental and environmental cues. These differentiated sublineages can orchestrate a broad range of effector activities during the initiation, expansion, and memory phase of an immune response. The contribution of CD4 cells to host defense against pathogenic invasion and regulation of autoimmunity is now well established. Emerging evidence suggests that CD4 cells also actively participate in shaping antitumor immunity. Here, we outline the biologic properties of CD4 T-cell subsets with an emphasis on their contribution to the antitumor response.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Authors' Affiliations: Department of Microbiology & Immunobiology, Division of Immunology, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
162
|
Tveita AA, Schjesvold F, Haabeth OA, Fauskanger M, Bogen B. Tumors Escape CD4+ T-cell–Mediated Immunosurveillance by Impairing the Ability of Infiltrating Macrophages to Indirectly Present Tumor Antigens. Cancer Res 2015; 75:3268-78. [DOI: 10.1158/0008-5472.can-14-3640] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/11/2015] [Indexed: 11/16/2022]
|
163
|
Tumor-specific Th2 responses inhibit growth of CT26 colon-cancer cells in mice via converting intratumor regulatory T cells to Th9 cells. Sci Rep 2015; 5:10665. [PMID: 26035423 PMCID: PMC4451845 DOI: 10.1038/srep10665] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/22/2015] [Indexed: 02/01/2023] Open
Abstract
The abnormality of immune regulation plays a critical role in the pathogenesis of cancer; the underlying mechanism has not been fully understood yet. This study aims to investigate the role of cancer specific T helper (Th)2 response in the inhibition of colon cancer (Cca) cell growth. The results showed that with Cca cell (CT26 cell) extracts as an antigen, the Cca-extract specific Th2 response was induced in the Cca-bearing mice. The Cca mass size was significantly reduced, or radically disappeared (5 out of 10; or 50%); the survival rate was markedly improved in mice immunized with Cca-extract, but not in those immunized with another tumor cell (U87 cell) extracts or to bovine serum albumin. The immunization with Cca-extract also induced Cca cell apoptosis and converted the intra-Cca Tregs to T helper (Th) 9 cells. In conclusion, Cca-specific Th2 responses inhibit Cca growth in a mouse model via inducing Cca cell apoptosis and converting intra-Cca Tregs to Th9 cells.
Collapse
|
164
|
Locke FL, Menges M, Veerapathran A, Coppola D, Gabrilovich D, Anasetti C. Survivin-specific CD4+ T cells are decreased in patients with survivin-positive myeloma. J Immunother Cancer 2015; 3:20. [PMID: 25992291 PMCID: PMC4437443 DOI: 10.1186/s40425-015-0065-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/28/2015] [Indexed: 11/10/2022] Open
Abstract
Background Survivin is a small protein inhibitor of apoptosis and a tumor associated antigen. Survivin expression in multiple myeloma is associated with poor prognosis, disease progression, and drug resistance. The CD4+ response against survivin remains uncharacterized. Methods In order to better understand the anti-tumor immune response to survivin, and optimize vaccination strategies, we characterized the spontaneous CD4+CD25- T cell response against survivin in healthy donors and myeloma patients using survivin derived peptide pools. Results Healthy donors and myeloma patients’ CD4+CD25- T cells exhibited a proliferative and IFN-gamma response against survivin peptides loaded onto autologous dendritic cells. We employed limiting dilution analysis to quantify the precursor frequency of survivin reactive CD4+CD25- T cells. Multiple myeloma patients (range 0% to 2.2x10-3%, n=12) had fewer survivin reactive CD4+CD25- T cells than healthy blood donors (range 1.1x10-3 to 8.4x10-3%, n=10), p = 0.021. The survivin reactive CD4+CD25- T cell precursor frequency was inversely associated with tumor survivin mRNA expression (p = 0.0028, r = −1.0, n = 6), and survivin tumor protein expression by IHC (p = 0.0295, r = −0.67, n = 10). A full length mutant survivin protein-pulsed dendritic cell vaccine expanded survivin reactive CD4+CD25- T cells after 12 days of in vitro culture (range 0-540x,median = 42x), and expansion was achieved even in patients with low baseline survivin reactive CD4+ precursors. Conclusions We have, for the first time, quantified the circulating CD4+CD25- precursor frequency against survivin and demonstrated this is lower in myeloma patients than healthy donors. The number of survivin reactive CD4+CD25- T cells is inversely associated with tumor survivin expression suggesting suppression of survivin responsive CD4+CD25- T cells. Further exploration of a full length mutant survivin protein vaccine which expands survivin reactive CD4+ cells independent of the survivin reactive precursor frequency is warranted. Electronic supplementary material The online version of this article (doi:10.1186/s40425-015-0065-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Frederick L Locke
- Moffitt Cancer Center, Department of Blood and Marrow Transplantation, Tampa, USA
| | - Meghan Menges
- Moffitt Cancer Center, Department of Blood and Marrow Transplantation, Tampa, USA
| | | | | | - Dmitry Gabrilovich
- The Wistar Institute, Tumor Microenvironment and Metastasis Program, Philadelphia, USA
| | - Claudio Anasetti
- Moffitt Cancer Center, Department of Blood and Marrow Transplantation, Tampa, USA
| |
Collapse
|
165
|
Wu TS, Wang LC, Liu SC, Hsu TY, Lin CY, Feng GJ, Chen JM, Liu HP, Chung IC, Yen TC, Chang YS, Liao SK, Chang C, Chow KPN. EBV oncogene N-LMP1 induces CD4 T cell-mediated angiogenic blockade in the murine tumor model. THE JOURNAL OF IMMUNOLOGY 2015; 194:4577-87. [PMID: 25847974 DOI: 10.4049/jimmunol.1400794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 02/23/2015] [Indexed: 01/01/2023]
Abstract
Antivascular immunity may provide long-term protection by preventing neovascularization that precedes tumor progression. Although the tumorigenesis promoted by EBV-encoded oncogene latent membrane protein 1 derived from Taiwanese nasopharyngeal carcinoma (N-LMP1) has been demonstrated, the potential of N-LMP1 for inducing immune surveillance remains elusive. In this article, we describe the immunogenicity of N-LMP1 (1510) and its induction of antivascular immunity in a transplantable tumor model in immunocompetent BALB/c mice. The immunogenicity of N-LMP1 was evaluated on the basis of tumor rejection following immunization. The impact of the immunization on the dynamics of tumor angiogenesis was assessed by temporal noninvasive dynamic contrast-enhanced magnetic resonance imaging and was further confirmed by histologic study and vascular count. Through the experiments of in vivo depletion and adoptive transfer, CD4 T cells were identified as effectors that depend on IFN-γ for tumor prevention. The response was further verified by the identification of an MHC H-2 I-E(d)-restricted peptide derived from N-LMP1 and by the immunization of mice with N-LMP1 peptide-loaded dendritic cells. These studies provide insight into N-LMP1-specific immunity in vivo, which suggests that CD4 T cells may play an important role in angiogenic surveillance against LMP1-associated cancer via tumor stroma targeting.
Collapse
Affiliation(s)
- Tzong-Shoon Wu
- Exploratory Research Laboratory, Development Center for Biotechnology, New Taipei City 221, Taiwan, Republic of China
| | - Lian-Chen Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China; Department of Parasitology, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan, Republic of China
| | - Shu-Chen Liu
- Molecular Medical Research Center, Chang Gung University, Taoyuan 333, Taiwan, Republic of China; Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan 320, Taiwan, Republic of China
| | - Ting-Yu Hsu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China
| | - Chun-Yen Lin
- Department of Hepatogastroenterology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, Republic of China
| | - Gou-Jin Feng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China
| | - Jian-Ming Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China
| | - Hao-Ping Liu
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan, Republic of China
| | - I-Che Chung
- Molecular Medical Research Center, Chang Gung University, Taoyuan 333, Taiwan, Republic of China
| | - Tzu-Chen Yen
- Department of Nuclear Medicine, Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, Republic of China
| | - Yu-Sun Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China; Molecular Medical Research Center, Chang Gung University, Taoyuan 333, Taiwan, Republic of China
| | - Shuen-Kuei Liao
- Ph.D. Program for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110, Taiwan, Republic of China
| | - Chen Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, Republic of China; and
| | - Kai-Ping N Chow
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China; Department of Microbiology and Immunology, Chang Gung University, Taoyuan 333, Taiwan, Republic of China
| |
Collapse
|
166
|
Chen JL, Dawoodji A, Tarlton A, Gnjatic S, Tajar A, Karydis I, Browning J, Pratap S, Verfaille C, Venhaus RR, Pan L, Altman DG, Cebon JS, Old LL, Nathan P, Ottensmeier C, Middleton M, Cerundolo V. NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int J Cancer 2015; 136:E590-601. [PMID: 25081390 DOI: 10.1002/ijc.29118] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/19/2014] [Accepted: 07/14/2014] [Indexed: 12/14/2022]
Abstract
Vaccination strategies based on repeated injections of NY-ESO-1 protein formulated in ISCOMATRIX particles (NY-ESO-1 ISCOMATRIX) have shown to elicit combined NY-ESO-1 specific antibody and T cell responses. However, it remains unclear whether heterologous prime-boost strategies based on the combination with NY-ESO-1 ISCOMATRIX with different NY-ESO-1 boosting reagents could be used to increase NY-ESO-1 CD8(+) or CD4(+) T cell responses. To address this question, we carried out a randomized clinical trial in 39 high-risk, resected melanoma patients vaccinated with NY-ESO-1 ISCOMATRIX, and then boosted with repeated injections of either recombinant fowlpox virus encoding full length NY-ESO-1 (rF-NY-ESO-1) (Arm A) or NY-ESO-1 ISCOMATRIX alone (Arm B). We have comprehensively analyzed NY-ESO-1 specific T cells and B cells response in all patients before and after vaccination for a total of seven time points per patient. NY-ESO-1 ISCOMATRIX alone elicited a strong NY-ESO-1 specific CD4(+) T cell and antibody response, which was maintained by both regiments at similar levels. However, CD8(+) T cell responses were significantly boosted in 3 out of 18 patients in Arm A after the first rF-NY-ESO-1 injection and such responses were maintained until the end of the trial, while no patients in Arm B showed similar CD8(+) T cell responses. In addition, our results clearly identified immunodominant regions in the NY-ESO-1 protein: NY-ESO-179-102 and NY-ESO-1115-138 for CD4+ T cells and NY-ESO-185-108 for CD8+ T cells in a large proportion of vaccinated patients. These regions of NY-ESO-1 protein should be considered in future clinical trials as immunodominant epitopes.
Collapse
Affiliation(s)
- Ji-Li Chen
- Radcliffe Department of Medicine, MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Flament H, Alonso Ramirez R, Prémel V, Joncker NT, Jacquet A, Scholl S, Lantz O. Modeling the specific CD4+ T cell response against a tumor neoantigen. THE JOURNAL OF IMMUNOLOGY 2015; 194:3501-12. [PMID: 25732731 DOI: 10.4049/jimmunol.1402405] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The antitumor activity of CD4(+) T cells is increasingly acknowledged in both humans and mice. The involved mechanisms have been mostly studied using transplanted tumor mouse systems. In these models, many tumor cells die at the time of implantation leading to the release of Ag in an inflammatory context contrasting with the slow and nondestructive growth of early-stage human tumors. In this study, we show that the presentation of a MHC class II-restricted model Ag (male, DBY) released by dying tumor cells may last more than 4 wk. The duration of Ag presentation varies according to the way the cells are killed before implantation. To avoid this artifactual early priming of the host precluding the study of the interactions between the immune system and tumors at the steady state, we generated a cell line expressing the DBY Ag in an inducible manner. Ag expression can be efficiently induced in vivo several days after tumor implantation. We show that the Ag reaches the lymph node and activates naive CD4(+) T cells to proliferate and recirculate. We did not observe de novo induction of tumor-specific regulatory T cells. However, we observed Th1/Th17 effector cells in the tumor draining lymph node and tumors. Thus, when a neoantigen appears in established tumors, the immune system is not ignorant and naive CD4(+) T cells are not tolerized. This opens up the possibility of therapeutic vaccines improving the immune response toward tumor-specific neoantigens.
Collapse
Affiliation(s)
| | | | | | | | | | - Suzy Scholl
- Center of Clinical Investigations in Biotherapy 1428 (Institut Gustave Roussy/Curie), Institut Curie, 75005 Paris, France; and Department of Medical Oncology, Institut Curie, 75005 Paris, France
| | - Olivier Lantz
- INSERM U932, Institut Curie, 75005 Paris, France; Center of Clinical Investigations in Biotherapy 1428 (Institut Gustave Roussy/Curie), Institut Curie, 75005 Paris, France; and
| |
Collapse
|
168
|
Zanetti M. Tapping CD4 T Cells for Cancer Immunotherapy: The Choice of Personalized Genomics. THE JOURNAL OF IMMUNOLOGY 2015; 194:2049-56. [DOI: 10.4049/jimmunol.1402669] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
169
|
Osborn JL, Greer SF. Metastatic melanoma cells evade immune detection by silencing STAT1. Int J Mol Sci 2015; 16:4343-61. [PMID: 25690042 PMCID: PMC4346960 DOI: 10.3390/ijms16024343] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/11/2015] [Indexed: 12/31/2022] Open
Abstract
Transcriptional activation of major histocompatibility complex (MHC) I and II molecules by the cytokine, interferon γ (IFN-γ), is a key step in cell-mediated immunity against pathogens and tumors. Recent evidence suggests that suppression of MHC I and II expression on multiple tumor types plays important roles in tumor immunoevasion. One such tumor is malignant melanoma, a leading cause of skin cancer-related deaths. Despite growing awareness of MHC expression defects, the molecular mechanisms by which melanoma cells suppress MHC and escape from immune-mediated elimination remain unknown. Here, we analyze the dysregulation of the Janus kinase (JAK)/STAT pathway and its role in the suppression of MHC II in melanoma cell lines at the radial growth phase (RGP), the vertical growth phase (VGP) and the metastatic phase (MET). While RGP and VGP cells both express MHC II, MET cells lack not only MHC II, but also the critical transcription factors, interferon response factor (IRF) 1 and its upstream activator, signal transducer and activator of transcription 1 (STAT1). Suppression of STAT1 in vitro was also observed in patient tumor samples, suggesting STAT1 silencing as a global mechanism of MHC II suppression and immunoevasion.
Collapse
Affiliation(s)
- JoDi Lynn Osborn
- Division of Cellular Biology and Immunology, Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| | - Susanna F Greer
- Division of Cellular Biology and Immunology, Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
170
|
Cheng Y, Luo Z, Yang S, Jia M, Zhao H, Xu W, Tang Y. The ratio of absolute lymphocyte count at interim of therapy to absolute lymphocyte count at diagnosis predicts survival in childhood B-lineage acute lymphoblastic leukemia. Leuk Res 2015; 39:144-50. [DOI: 10.1016/j.leukres.2014.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/01/2014] [Accepted: 11/22/2014] [Indexed: 01/17/2023]
|
171
|
Lu X, Ding ZC, Cao Y, Liu C, Habtetsion T, Yu M, Lemos H, Salman H, Xu H, Mellor AL, Zhou G. Alkylating agent melphalan augments the efficacy of adoptive immunotherapy using tumor-specific CD4+ T cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:2011-21. [PMID: 25560408 DOI: 10.4049/jimmunol.1401894] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In recent years, the immune-potentiating effects of some widely used chemotherapeutic agents have been increasingly appreciated. This provides a rationale for combining conventional chemotherapy with immunotherapy strategies to achieve durable therapeutic benefits. Previous studies have implicated the immunomodulatory effects of melphalan, an alkylating agent commonly used to treat multiple myeloma, but the underlying mechanisms remain obscure. In the present study, we investigated the impact of melphalan on endogenous immune cells as well as adoptively transferred tumor-specific CD4(+) T cells in tumor-bearing mice. We showed that melphalan treatment resulted in a rapid burst of inflammatory cytokines and chemokines during the cellular recovery phase after melphalan-induced myelodepletion and leukodepletion. After melphalan treatment, tumor cells exhibited characteristics of immunogenic cell death, including membrane translocation of the endoplasmic reticulum-resident calreticulin and extracellular release of high-mobility group box 1. Additionally, there was enhanced tumor Ag uptake by dendritic cells in the tumor-draining lymph node. Consistent with these immunomodulatory effects, melphalan treatment of tumor-bearing mice led to the activation of the endogenous CD8(+) T cells and, more importantly, effectively drove the clonal expansion and effector differentiation of adoptively transferred tumor-specific CD4(+) T cells. Notably, the combination of melphalan and CD4(+) T cell adoptive cell therapy was more efficacious than either treatment alone in prolonging the survival of mice with advanced B cell lymphomas or colorectal tumors. These findings provide mechanistic insights into melphalan's immunostimulatory effects and demonstrate the therapeutic potential of combining melphalan with adoptive cell therapy utilizing antitumor CD4(+) T cells.
Collapse
Affiliation(s)
- Xiaoyun Lu
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; Division of Digestive Endoscopy, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Zhi-Chun Ding
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912
| | - Yang Cao
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Chufeng Liu
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; Department of Orthodontics, Guangdong Provincial Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Tsadik Habtetsion
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912
| | - Miao Yu
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912
| | - Henrique Lemos
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912
| | - Huda Salman
- Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; and
| | - Hongyan Xu
- Department of Biostatistics and Epidemiology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912
| | - Andrew L Mellor
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; and
| | - Gang Zhou
- Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912; and
| |
Collapse
|
172
|
Brisson L, Pouyet L, N’guessan P, Garcia S, Lopes N, Warcollier G, Iovanna J, Carrier A. The Thymus-Specific Serine Protease TSSP/PRSS16 Is Crucial for the Antitumoral Role of CD4+ T Cells. Cell Rep 2015; 10:39-46. [DOI: 10.1016/j.celrep.2014.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 10/30/2014] [Accepted: 12/04/2014] [Indexed: 01/20/2023] Open
|
173
|
Tveita AA, Haabeth OA, Bogen B. Limitations of bystander killing in Th1/M1 immune responses against a secreted tumor antigen. Oncoimmunology 2014; 3:e954953. [PMID: 25941607 DOI: 10.4161/21624011.2014.954953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 07/19/2014] [Indexed: 11/19/2022] Open
Abstract
T-cell recognition of tumor antigens presented on tumor-infiltrating macrophages (TAMs) induces a tumoricidal M1-like phenotype. Resultant indirect immune responses could eliminate not only antigen secreting (AgPOS), but also antigen negative (AgNEG) tumor cells via bystander killing. Such broad-spectrum response could eliminate antigenically heterogeneous tumors. Using an in vivo model of CD4+ T-cell mediated immunity against MHC II negative myeloma cells, bystander killing of AgNEG cells was ineffective due to strict spatial constraints of Th1-induced TAM cytotoxicity.
Collapse
Key Words
- ACT, Adoptive cell therapy
- APC, Antigen-presenting cell
- Ag, Antigen
- CD4+ T cell response
- IFN, Interferon gamma
- Id, idiotype
- LPS, Lipopolysaccharide
- NK, Natural killer
- SCID, Severe combined immunodeficiency
- TAM, Tumor-associated macrophage
- TCR-Tg, T-cell receptor transgenic
- bystander killing
- macrophage cytotoxicity
- multiple myeloma
Collapse
Affiliation(s)
- Anders Aune Tveita
- Centre for Immune Regulation; Institute of Immunology; University of Oslo and Oslo University Hospital ; Oslo, Norway
| | - Ole Audun Haabeth
- Centre for Immune Regulation; Institute of Immunology; University of Oslo and Oslo University Hospital ; Oslo, Norway
| | - Bjarne Bogen
- Centre for Immune Regulation; Institute of Immunology; University of Oslo and Oslo University Hospital ; Oslo, Norway ; KG Jebsen Centre for Research on Influenza Vaccines; Institute of Immunology; University of Oslo and Oslo University Hospital ; Oslo, Norway
| |
Collapse
|
174
|
Gardyan A, Osen W, Zörnig I, Podola L, Agarwal M, Aulmann S, Ruggiero E, Schmidt M, Halama N, Leuchs B, von Kalle C, Beckhove P, Schneeweiss A, Jäger D, Eichmüller SB. Identification of NY-BR-1-specific CD4(+) T cell epitopes using HLA-transgenic mice. Int J Cancer 2014; 136:2588-97. [PMID: 25387692 DOI: 10.1002/ijc.29322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/21/2014] [Indexed: 12/11/2022]
Abstract
Breast cancer represents the second most common cancer type worldwide and has remained the leading cause of cancer-related deaths among women. The differentiation antigen NY-BR-1 appears overexpressed in invasive mammary carcinomas compared to healthy breast tissue, thus representing a promising target antigen for T cell based tumor immunotherapy approaches. Since efficient immune attack of tumors depends on the activity of tumor antigen-specific CD4(+) effector T cells, NY-BR-1 was screened for the presence of HLA-restricted CD4(+) T cell epitopes that could be included in immunological treatment approaches. Upon NY-BR-1-specific DNA immunization of HLA-transgenic mice and functional ex vivo analysis, a panel of NY-BR-1-derived library peptides was determined that specifically stimulated IFNγ secretion among splenocytes of immunized mice. Following in silico analyses, four candidate epitopes were determined which were successfully used for peptide immunization to establish NY-BR-1-specific, HLA-DRB1*0301- or HLA-DRB1*0401-restricted CD4(+) T cell lines from splenocytes of peptide immunized HLA-transgenic mice. Notably, all four CD4(+) T cell lines recognized human HLA-DR-matched dendritic cells (DC) pulsed with lysates of NY-BR-1 expressing human tumor cells, demonstrating natural processing of these epitopes also within the human system. Finally, CD4(+) T cells specific for all four CD4(+) T cell epitopes were detectable among PBMC of breast cancer patients, showing that CD4(+) T cell responses against the new epitopes are not deleted nor inactivated by self-tolerance mechanisms. Our results present the first NY-BR-1-specific HLA-DRB1*0301- and HLA-DRB1*0401-restricted T cell epitopes that could be exploited for therapeutic intervention against breast cancer.
Collapse
Affiliation(s)
- Adriane Gardyan
- Department of Translational Immunology, German Cancer Research Center Heidelberg (DKFZ), Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Binsfeld M, Beguin Y, Belle L, Otjacques E, Hannon M, Briquet A, Heusschen R, Drion P, Zilberberg J, Bogen B, Baron F, Caers J. Establishment of a murine graft-versus-myeloma model using allogeneic stem cell transplantation. PLoS One 2014; 9:e113764. [PMID: 25415267 PMCID: PMC4240591 DOI: 10.1371/journal.pone.0113764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 10/29/2014] [Indexed: 11/18/2022] Open
Abstract
Background Multiple myeloma (MM) is a malignant plasma cell disorder with poor long-term survival and high recurrence rates. Despite evidence of graft-versus-myeloma (GvM) effects, the use of allogeneic hematopoietic stem cell transplantation (allo-SCT) remains controversial in MM. In the current study, we investigated the anti-myeloma effects of allo-SCT from B10.D2 mice into MHC-matched myeloma-bearing Balb/cJ mice, with concomitant development of chronic graft-versus-host disease (GvHD). Methods and results Balb/cJ mice were injected intravenously with luciferase-transfected MOPC315.BM cells, and received an allogeneic (B10.D2 donor) or autologous (Balb/cJ donor) transplant 30 days later. We observed a GvM effect in 94% of the allogeneic transplanted mice, as the luciferase signal completely disappeared after transplantation, whereas all the autologous transplanted mice showed myeloma progression. Lower serum paraprotein levels and lower myeloma infiltration in bone marrow and spleen in the allogeneic setting confirmed the observed GvM effect. In addition, the treated mice also displayed chronic GvHD symptoms. In vivo and in vitro data suggested the involvement of effector memory CD4 and CD8 T cells associated with the GvM response. The essential role of CD8 T cells was demonstrated in vivo where CD8 T-cell depletion of the graft resulted in reduced GvM effects. Finally, TCR Vβ spectratyping analysis identified Vβ families within CD4 and CD8 T cells, which were associated with both GvM effects and GvHD, whereas other Vβ families within CD4 T cells were associated exclusively with either GvM or GvHD responses. Conclusions We successfully established an immunocompetent murine model of graft-versus-myeloma. This is the first murine GvM model using immunocompetent mice that develop MM which closely resembles human MM disease and that are treated after disease establishment with an allo-SCT. Importantly, using TCR Vβ spectratyping, we also demonstrated the presence of GvM unique responses potentially associated with the curative capacity of this immunotherapeutic approach.
Collapse
Affiliation(s)
- Marilène Binsfeld
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Ludovic Belle
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Eléonore Otjacques
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Muriel Hannon
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Alexandra Briquet
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Roy Heusschen
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | | | - Jenny Zilberberg
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey, United States of America
| | - Bjarne Bogen
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- KG Jebsen centre for research on influenza vaccines, Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
176
|
Microenvironment of tumor-draining lymph nodes: opportunities for liposome-based targeted therapy. Int J Mol Sci 2014; 15:20209-39. [PMID: 25380524 PMCID: PMC4264163 DOI: 10.3390/ijms151120209] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 02/07/2023] Open
Abstract
The World Health Organization (WHO) recently reported that the total number of global cancer cases in 2013 reached 14 million, a 10% rise since 2008, while the total number of cancer deaths reached 8.2 million, a 5.2% increase since 2008. Metastasis is the major cause of death from cancer, accounting for 90% of all cancer related deaths. Tumor-draining lymph nodes (TDLN), the sentinel nodes, are the first organs of metastasis in several types of cancers. The extent of metastasis in the TDLN is often used in disease staging and prognosis evaluation in cancer patients. Here, we describe the microenvironment of the TDLN and review the recent literature on liposome-based therapies directed to immune cells within the TDLN with the intent to target cancer cells.
Collapse
|
177
|
Lorvik KB, Haabeth OAW, Clancy T, Bogen B, Corthay A. Molecular profiling of tumor-specific T H1 cells activated in vivo. Oncoimmunology 2014; 2:e24383. [PMID: 23762808 PMCID: PMC3667914 DOI: 10.4161/onci.24383] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 12/21/2022] Open
Abstract
The central role of tumor-specific TH1 cells in anticancer immune responses is becoming increasingly appreciated. However, little is known about how these cells are generated in vivo. Here, we used flow cytometry and gene expression microarrays to characterize the primary activation and TH1 differentiation of naïve tumor-specific CD4+ T cells in a mouse model of cancer immunosurveillance. We took advantage of T-cell receptor-transgenic mice in which CD4+ T cells recognize a tumor-specific antigen secreted by MHC class II-negative MOPC315 myeloma cells. Cancer cells were injected subcutaneously and T-cell activation was analyzed in draining lymph nodes and at the incipient tumor site 8 d later. Upon activation and migration to incipient tumor sites, tumor-specific CD4+ T cells exhibited the upregulation of 29 cell-surface molecules (CD2, CD5, CD11a, CD18, CD25, CD28, CD44, CD45, CD49d, CD51, CD54, CD69, CD71, CD83, CD86, CD90, CD95, CD102, CD122, CD153, CD166, CD200, CD249, CD254, CD274, CD279, Ly6C, MHC class I and CCR7) and the downregulation of five (CD27, CD31, CD45RB, CD62L and CD126). Activated CD4+ T cells produced interferon γ, a cytokine consistent with a TH1-polarized response, tumor necrosis factor α as well as interleukin (IL)-2, IL-3 and IL-10. The activation of naïve tumor-specific CD4+ T cells in draining lymph nodes resulted in the upregulation of 609 genes and the downregulation of 284 genes. The bioinformatic analysis of differentially expressed genes identified functional pathways related to tumor-specific TH1 cell activation. This study may represent a useful resource to guide the development of TH1-based immunotherapies against cancer.
Collapse
Affiliation(s)
- Kristina Berg Lorvik
- Centre for Immune Regulation; Department of Immunology; Oslo University Hospital Rikshospitalet and University of Oslo; Oslo, Norway
| | | | | | | | | |
Collapse
|
178
|
Spear P, Barber A, Sentman CL. Collaboration of chimeric antigen receptor (CAR)-expressing T cells and host T cells for optimal elimination of established ovarian tumors. Oncoimmunology 2014; 2:e23564. [PMID: 23734311 PMCID: PMC3654581 DOI: 10.4161/onci.23564] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/07/2013] [Accepted: 01/09/2013] [Indexed: 12/24/2022] Open
Abstract
Conditioning strategies that deplete host lymphocytes have been shown to enhance clinical responses to some adoptive T-cell therapies. However, host T cells are capable of eliminating tumor cells upon the relief of immunosuppression, indicating that lymphodepletion prior to T-cell transfer may reduce optimal tumor protection elicited by cell treatments that are capable of shaping host immunity. In this study, we show that adoptively transferred T cells bearing a chimeric antigen receptor (CAR) harness endogenous T cells for optimal tumor elimination and the development of a tumor-specific memory T cell response. Mice bearing ID8 ovarian cancer cells were treated with T cells transduced with a NKG2D-based CAR. CAR-expressing T cells increased the number of host CD4+ and CD8+ T cells at the tumor site in a CXCR3-dependent manner and increased the number of antigen-specific host CD4+ T cells in the tumor and draining lymph nodes. In addition, the administration of CAR-expressing T cells increased antigen presentation to CD4+ T cells, and this increase was dependent on interferon γ and granulocyte-macrophage colony-stimulating factor produced by the former. Host CD4+ T cells were sufficient for optimal tumor protection mediated by NKG2D CAR-expressing T cells, but they were not necessary if CD4+ T cells were adoptively co-transferred. However, host CD4+ T cells were essential for the development of an antigen-specific memory T-cell response to tumor cells. Moreover, optimal tumor elimination as orchestrated by NKG2D CAR-expressing T cells was dependent on host CD8+ T cells. These results demonstrate that adoptively transferred T cells recruit and activate endogenous T-cell immunity to enhance the elimination of tumor cells and the development of tumor-specific memory responses.
Collapse
Affiliation(s)
- Paul Spear
- Department of Microbiology & Immunology; The Geisel School of Medicine at Dartmouth; Lebanon, NH USA
| | | | | |
Collapse
|
179
|
Haabeth OAW, Bogen B, Corthay A. A model for cancer-suppressive inflammation. Oncoimmunology 2014; 1:1146-1155. [PMID: 23170261 PMCID: PMC3494627 DOI: 10.4161/onci.21542] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In oncology, inflammation is generally regarded as a cancer-promoting process only. Here, we argue that this view may represent a misleading oversimplification. We present evidence from our own work and from the literature documenting cancer-suppressive aspects of inflammation. We propose that specific types of inflammation, in particular inflammation driven by tumor-specific Th1 cells, may repress rather than promote cancer. Th1 cells collaborate with tumor-infiltrating M1 macrophages to efficiently recognize and eliminate malignant cells. In a Th1 environment, pro-inflammatory cytokines such as interleukin (IL)-1α, IL-1β, IL-6 and tumor-necrosis factor α (TNFα) enhance anti-cancer immunity. Inducing Th1-type inflammation may significantly improve immunotherapeutic strategies against cancer.
Collapse
Affiliation(s)
- Ole Audun Werner Haabeth
- Centre for Immune Regulation, Department of Immunology; Oslo University Hospital Rikshospitalet and University of Oslo; Oslo, Norway
| | | | | |
Collapse
|
180
|
Narwani V, Gabriel J, Boyd K, Chevassut T. Absolute lymphocyte count at day 29 of treatment is a powerful predictor of outcome in multiple myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2014; 15:222-6. [PMID: 25468320 DOI: 10.1016/j.clml.2014.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/09/2014] [Accepted: 10/20/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Survival outcomes for patients who receive treatment for newly diagnosed multiple myeloma (MM) are highly variable. PATIENTS AND METHODS We conducted a retrospective analysis of 38 unselected MM patients who received treatment with cyclophosphamide, thalidomide, and dexamethasone to evaluate the prognostic value of the absolute lymphocyte count at diagnosis and at the end of the initial cycle of treatment defined as day 29, termed ALC-29. The median follow-up was 54 months (range, 2-83 months). RESULTS We found that ALC-29, as a continuous variable, was a predictor of overall survival (OS) in MM patients (hazard ratio, 0.208; 95% confidence interval, 0.093-0.689; P = .007). Patients with an ALC-29 ≥ 0.8 × 10(9)/L (n = 16) experienced a superior median OS compared with patients with an ALC-29 < 0.8 × 10(9)/L (n = 22) with a median OS of 58.3 months versus 42.5 months respectively (P = .006). Multivariate analysis confirmed that ALC-29 ≥ 0.8 × 10(9)/L was an independent prognostic indicator of OS in our cohort of MM patients. CONCLUSION We concluded that the ALC-29 is a useful and simple predictor of outcome in newly diagnosed MM patients who receive standard chemotherapy. Our results support the hypothesis that host immunity plays an important role in tumor control in MM.
Collapse
Affiliation(s)
- Vishal Narwani
- Brighton and Sussex Medical School, Medical Research Building, University of Sussex, Brighton, United Kingdom
| | - Joseph Gabriel
- Brighton and Sussex Medical School, Medical Research Building, University of Sussex, Brighton, United Kingdom
| | - Kevin Boyd
- Royal Sussex County Hospital, Eastern Road, Brighton, United Kingdom
| | - Timothy Chevassut
- Brighton and Sussex Medical School, Medical Research Building, University of Sussex, Brighton, United Kingdom; Royal Sussex County Hospital, Eastern Road, Brighton, United Kingdom.
| |
Collapse
|
181
|
Noyan F, Lieke T, Taubert R, Sievers M, Dywicki J, Hapke M, Falk CS, Manns MP, Jaeckel E, Hardtke-Wolenski M. Naive tumour-specific CD4+ T cells were efficiently primed in acute lymphoblastic leukaemia. Scand J Immunol 2014; 80:161-8. [PMID: 24910003 DOI: 10.1111/sji.12198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/21/2014] [Indexed: 12/31/2022]
Abstract
The recognition and neutralization of tumour cells is one of the big challenges in immunity. The immune system has to recognize syngeneic tumour cells and has to be primed and respond in an adequate manner. Priming of a leukaemia-specific immune response is a crucial step in tumour immunology that can mislead to tumour tolerance either by T cell ignorance, deletion or Treg induction. To resemble the situation of acute lymphoblastic leukaemia (ALL) in patients, we used the murine BALB/c model with syngeneic BM185 tumour cells. We established a tumour cell line that expresses the neo-antigen ovalbumin (BM185-OVA/GFP) to allow the application of T cell receptor transgenic, antigen-specific CD4(+) T cells. Here, we demonstrate that effective anti-ALL immunity can be established by in vivo priming of CD4(+) T cells that is sufficient to differentiate into effector cells. Yet they failed to control tumour alone, but initiated a Th1 response. An efficient tumour clearance was dependent on both antigen-specific CD4(+) T cells and CD8(+) effector T cells from the endogenous repertoire. The tolerogeneic milieu was characterized by increased Tregs numbers and elevated IL-10 level. Tregs hamper effective antitumour immune response, but their depletion did not result in reduced tumour growth. In contrast, neutralization of IL-10 improved median mouse survival. Future therapies should focus on establishing a strong CD4+ T cells response, either by adjuvant or by adoptive transfer.
Collapse
Affiliation(s)
- F Noyan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Laoui D, Van Overmeire E, De Baetselier P, Van Ginderachter JA, Raes G. Functional Relationship between Tumor-Associated Macrophages and Macrophage Colony-Stimulating Factor as Contributors to Cancer Progression. Front Immunol 2014; 5:489. [PMID: 25339957 PMCID: PMC4188035 DOI: 10.3389/fimmu.2014.00489] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/22/2014] [Indexed: 12/14/2022] Open
Abstract
The current review article describes the functional relationship between tumor-associated macrophages (TAM) as key cellular contributors to cancer malignancy on the one hand and macrophage-colony-stimulating factor (M-CSF or CSF-1) as an important molecular contributor on the other. We recapitulate the available data on expression of M-CSF and the M-CSF receptor (M-CSFR) in human tumor tissue as constituents of a stromal macrophage signature and on the limits of the predictive and prognostic value of plasma M-CSF levels. After providing an update on current insights into the nature of TAM heterogeneity at the level of M1/M2 phenotype and TAM subsets, we give an overview of experimental evidence, based on genetic, antibody-mediated, and pharmacological disruption of M-CSF/M-CSFR signaling, for the extent to which M-CSFR signaling can not only determine the TAM quantity, but can also contribute to shaping the phenotype and heterogeneity of TAM and other related tumor-infiltrating myeloid cells (TIM). Finally, we review the accumulating information on the – sometimes conflicting – effects blocking M-CSFR signaling may have on various aspects of cancer progression such as tumor growth, invasion, angiogenesis, metastasis, and resistance to therapy and we thereby discuss in how far these different effects actually reflect a contribution of TAM.
Collapse
Affiliation(s)
- Damya Laoui
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Eva Van Overmeire
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Patrick De Baetselier
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Geert Raes
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| |
Collapse
|
183
|
Erin N, Podnos A, Tanriover G, Duymuş Ö, Cote E, Khatri I, Gorczynski RM. Bidirectional effect of CD200 on breast cancer development and metastasis, with ultimate outcome determined by tumor aggressiveness and a cancer-induced inflammatory response. Oncogene 2014; 34:3860-70. [PMID: 25263452 DOI: 10.1038/onc.2014.317] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 08/02/2014] [Accepted: 08/09/2014] [Indexed: 12/27/2022]
Abstract
CD200 acts through its receptor (CD200R) to inhibit excessive inflammation. The role of CD200-CD200R1 interaction in tumor immunity is poorly understood. In this study, we examined the role of CD200-CD200R1 interaction in the progression and metastasis of highly aggressive 4THM murine-breast carcinoma using CD200 transgenic (CD200(tg)) and CD200R1 knock-out (CD200R1(-)(/-)) BALB/c mice. 4THM cells induce extensive visceral metastasis and neutrophil infiltration in affected tissues. CD200 overexpression in the host was associated with decreased primary tumor growth and metastasis, whereas lack of CD200R1 expression by host cells was associated with enhanced visceral metastasis. Absence of CD200R1 expression led to decreased tumor-infiltrating-cytotoxic T cells and increased the release of inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin (IL)-6. In contrast, CD200 overexpression led to increased tumor-induced interferon-γ and IL-10 response and decreased TNF-α and IL-6 release. Neutrophil infiltration of tissues was markedly decreased in CD200(tg) animals and increased in CD200R1(-/-) mice. These findings are contradictory to what has been reported in the EMT6 mouse breast-cancer model. Other distinguishing features of tumor elicited by EMT6 and 4THM cell injections were also examined. Visceral tissues from mice bearing EMT6 tumors showed a lack of neutrophil infiltration and decreased IL-6 release in CD200R1(-/-) mice. EMT6 and 4THM cells also differed in vimentin expression and in vitro migration rate, which was markedly lower in EMT6 tumors. These results support the hypothesis that CD200 expression can alter immune responses, and can inhibit metastatic growth of tumor cells that induce systemic and local inflammatory response. Increasing CD200 activity/signaling might be an important therapeutic strategy for treatment of aggressive breast carcinomas.
Collapse
Affiliation(s)
- N Erin
- Department of Medical Pharmacology, School of Medicine, Akdeniz University, Antalya City, Antalya, Turkey
| | - A Podnos
- University Health Network, Toronto General Hospital, Toronto, ON, Canada
| | - G Tanriover
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Antalya, Turkey
| | - Ö Duymuş
- Department of Medical Pharmacology, School of Medicine, Akdeniz University, Antalya City, Antalya, Turkey
| | - E Cote
- University Health Network, Toronto General Hospital, Toronto, ON, Canada
| | - I Khatri
- University Health Network, Toronto General Hospital, Toronto, ON, Canada
| | - R M Gorczynski
- University Health Network, Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
184
|
Murine Splenic CD4+ T Cells, Induced by Innate Immune Cell Interactions and Secreted Factors, Develop Antileukemia Cytotoxicity. Cancer Immunol Res 2014; 2:1113-24. [DOI: 10.1158/2326-6066.cir-13-0208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
185
|
Aspord C, Leloup C, Reche S, Plumas J. pDCs efficiently process synthetic long peptides to induce functional virus- and tumour-specific T-cell responses. Eur J Immunol 2014; 44:2880-92. [DOI: 10.1002/eji.201444588] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/12/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Caroline Aspord
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| | - Claire Leloup
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| | - Sabine Reche
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| | - Joel Plumas
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| |
Collapse
|
186
|
Kasamatsu J, Takahashi S, Azuma M, Matsumoto M, Morii-Sakai A, Imamura M, Teshima T, Takahashi A, Hirohashi Y, Torigoe T, Sato N, Seya T. PolyI:C and mouse survivin artificially embedding human 2B peptide induce a CD4+ T cell response to autologous survivin in HLA-A*2402 transgenic mice. Immunobiology 2014; 220:74-82. [PMID: 25257859 DOI: 10.1016/j.imbio.2014.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/04/2014] [Accepted: 08/06/2014] [Indexed: 11/29/2022]
Abstract
CD4(+) T cell effectors are crucial for establishing antitumor immunity. Dendritic cell maturation by immune adjuvants appears to facilitate subset-specific CD4(+) T cell proliferation, but the adjuvant effect for CD4 T on induction of cytotoxic T lymphocytes (CTLs) is largely unknown. Self-antigenic determinants with low avidity are usually CD4 epitopes in mutated proteins with tumor-associated class I-antigens (TAAs). In this study, we made a chimeric version of survivin, a target of human CTLs. The chimeric survivin, where human survivin-2B containing a TAA was embedded in the mouse survivin frame (MmSVN2B), was used to immunize HLA-A-2402/K(b)-transgenic (HLA24(b)-Tg) mice. Subcutaneous administration of MmSVN2B or xenogeneic human survivin (control HsSNV2B) to HLA24(b)-Tg mice failed to induce an immune response without co-administration of an RNA adjuvant polyI:C, which was required for effector induction in vivo. Although HLA-A-2402/K(b) presented the survivin-2B peptide in C57BL/6 mice, 2B-specific tetramer assays showed that no CD8(+) T CTLs specific to survivin-2B proliferated above the detection limit in immunized mice, even with polyI:C treatment. However, the CD4(+) T cell response, as monitored by IFN-γ, was significantly increased in mice given polyI:C+MmSVN2B. The Th1 response and antibody production were enhanced in the mice with polyI:C. The CD4 epitope responsible for effector function was not Hs/MmSNV13-27, a nonconserved region between human and mouse survivin, but region 53-67, which was identical between human and mouse survivin. These results suggest that activated, self-reactive CD4(+) helper T cells proliferate in MmSVN2B+polyI:C immunization and contribute to Th1 polarization followed by antibody production, but hardly participate in CTL induction.
Collapse
Affiliation(s)
- Jun Kasamatsu
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan
| | - Shojiro Takahashi
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan; Department of Hematology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan
| | - Masahiro Azuma
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan
| | - Misako Matsumoto
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan
| | - Akiko Morii-Sakai
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan
| | - Masahiro Imamura
- Department of Hematology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan
| | - Akari Takahashi
- Department of Pathology, Sapporo Medical University School of Medicine, Chuoh-ku, Sapporo, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, Chuoh-ku, Sapporo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Chuoh-ku, Sapporo, Japan
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Chuoh-ku, Sapporo, Japan
| | - Tsukasa Seya
- Department of Microbiology and Immunology, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo, Japan.
| |
Collapse
|
187
|
Cellular immunotherapy in multiple myeloma: lessons from preclinical models. Biochim Biophys Acta Rev Cancer 2014; 1846:392-404. [PMID: 25109893 DOI: 10.1016/j.bbcan.2014.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 12/20/2022]
Abstract
The majority of multiple myeloma patients relapse with the current treatment strategies, raising the need for alternative therapeutic approaches. Cellular immunotherapy is a rapidly evolving field and currently being translated into clinical trials with encouraging results in several cancer types, including multiple myeloma. Murine multiple myeloma models are of critical importance for the development and refinement of cellular immunotherapy. In this review, we summarize the immune cell changes that occur in multiple myeloma patients and we discuss the cell-based immunotherapies that have been tested in multiple myeloma, with a focus on murine models.
Collapse
|
188
|
Tveita AA, Schjesvold FH, Sundnes O, Haabeth OAW, Haraldsen G, Bogen B. Indirect CD4+ T-cell-mediated elimination of MHC II(NEG) tumor cells is spatially restricted and fails to prevent escape of antigen-negative cells. Eur J Immunol 2014; 44:2625-37. [PMID: 24846412 DOI: 10.1002/eji.201444659] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 04/28/2014] [Accepted: 05/16/2014] [Indexed: 11/06/2022]
Abstract
Tumor-specific Th1 cells can activate tumor-infiltrating macrophages that eliminate MHC class II negative (MHC II(NEG)) tumor cells. Activated M1-like macrophages lack antigen (Ag) receptors, and are presumably unable to discriminate and thus kill both Ag-positive (Ag(POS)) and Ag-negative (Ag(NEG)) tumor cells (bystander killing). The lack of specificity of macrophage-mediated cytotoxicity might be of clinical importance as it could provide a means of avoiding tumor escape. Here, we have tested this idea using mixed populations of Ag(POS) and Ag(NEG) tumor cells in a TCR-transgenic model in which CD4(+) T cells recognize a secreted tumor-specific antigen. Surprisingly, while Ag(POS) tumor cells were recognized and rejected, Ag(NEG) cells grew unimpeded and formed tumors. We further demonstrated that macrophage-mediated cytotoxicity was spatially restricted to areas dominated by Ag(POS) tumor cells, sparing Ag(NEG) tumor cells in the vicinity. As a consequence, macrophage tumoricidal activity did not confer bystander killing in vivo. The present results offer novel insight into the mechanisms of indirect Th1-mediated elimination of MHC II(NEG) tumor cells.
Collapse
Affiliation(s)
- Anders A Tveita
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
189
|
Kreutzman A, Ilander M, Porkka K, Vakkila J, Mustjoki S. Dasatinib promotes Th1-type responses in granzyme B expressing T-cells. Oncoimmunology 2014; 3:e28925. [PMID: 25083322 PMCID: PMC4106168 DOI: 10.4161/onci.28925] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/10/2014] [Accepted: 04/16/2014] [Indexed: 02/08/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have dramatically improved the outcome of chronic myeloid leukemia (CML). Besides inhibiting target kinases in leukemic cells, 2nd generation TKI dasatinib also inhibits off-targets in immune effector cells resulting in atypical immune responses in some patients. Dasatinib has been described to increase the proportion of late effector memory T-cells, however, to date no follow-up studies have been performed in first-line patients. In this study, we explored the functional properties of T-cells using primary samples from CML patients (n = 28) on TKI therapy. Granzyme B (GrB) was used as a marker for late phase antigen experienced CD4+ and CD8+ T-cells. Dasatinib treatment increased the numbers of both GrB expressing memory CD4+ and CD8+ T-cells when compared with healthy controls. Functionally, the GrB+CD4+ T-cells were highly active and differentiated into Th1-type T-cells capable of producing IFN-γ, which is important for tumor control. Similar kind of increase was not observed during imatinib or nilotinib therapy. These data support the dual mode of action of dasatinib: potent BCR-ABL1 inhibition in leukemic cells is accompanied by the enhancement of cellular immunity, which may have implications in the long-term control of leukemia.
Collapse
Affiliation(s)
- Anna Kreutzman
- Hematology Research Unit Helsinki; University of Helsinki; Helsinki, Finland ; Department of Hematology; Helsinki University Central Hospital Cancer Center; Helsinki, Finland
| | - Mette Ilander
- Hematology Research Unit Helsinki; University of Helsinki; Helsinki, Finland ; Department of Hematology; Helsinki University Central Hospital Cancer Center; Helsinki, Finland
| | - Kimmo Porkka
- Hematology Research Unit Helsinki; University of Helsinki; Helsinki, Finland ; Department of Hematology; Helsinki University Central Hospital Cancer Center; Helsinki, Finland
| | - Jukka Vakkila
- Hematology Research Unit Helsinki; University of Helsinki; Helsinki, Finland ; Department of Hematology; Helsinki University Central Hospital Cancer Center; Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki; University of Helsinki; Helsinki, Finland ; Department of Hematology; Helsinki University Central Hospital Cancer Center; Helsinki, Finland
| |
Collapse
|
190
|
Engelbertsen D, Rattik S, Knutsson A, Björkbacka H, Bengtsson E, Nilsson J. Induction of T helper 2 responses against human apolipoprotein B100 does not affect atherosclerosis in ApoE−/− mice. Cardiovasc Res 2014; 103:304-12. [DOI: 10.1093/cvr/cvu131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
191
|
Gérard C, Baudson N, Ory T, Louahed J. Tumor mouse model confirms MAGE-A3 cancer immunotherapeutic as an efficient inducer of long-lasting anti-tumoral responses. PLoS One 2014; 9:e94883. [PMID: 24830315 PMCID: PMC4022504 DOI: 10.1371/journal.pone.0094883] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/20/2014] [Indexed: 11/18/2022] Open
Abstract
Purpose MAGE-A3 is a potential target for immunotherapy due to its tumor-specific nature and expression in several tumor types. Clinical data on MAGE-A3 immunotherapy have raised many questions that can only be addressed by using animal models. In the present study, different aspects of the murine anti-tumor immune responses induced by a recombinant MAGE-A3 protein (recMAGE-A3) in combination with different immunostimulants (AS01, AS02, CpG7909 or AS15) were investigated. Experimental Design and Results Based on cytokine profile analyses and protection against challenge with MAGE-A3-expressing tumor, the combination recMAGE-A3+AS15 was selected for further experimental work, in particular to study the mechanisms of anti-tumor responses. By using MHC class I-, MHC class II-, perforin-, B-cell- and IFN-γ- knock-out mice and CD4+ T cell-, CD8+ T cell- and NK cell- depleted mice, we demonstrated that CD4+ T cells and NK cells are the main anti-tumor effectors, and that IFN-γ is a major effector molecule. This mouse tumor model also established the need to repeat recMAGE-A3+AS15 injections to sustain efficient anti-tumor responses. Furthermore, our results indicated that the efficacy of tumor rejection by the elicited anti-MAGE-A3 responses depends on the proportion of tumor cells expressing MAGE-A3. Conclusions The recMAGE-A3+AS15 cancer immunotherapy efficiently induced an antigen-specific, functional and long-lasting immune response able to recognize and eliminate MAGE-A3-expressing tumor cells up to several months after the last immunization in mice. The data highlighted the importance of the immunostimulant to induce a Th1-type immune response, as well as the key role played by IFN-γ, CD4+ T cells and NK cells in the anti-tumoral effect.
Collapse
Affiliation(s)
| | | | - Thierry Ory
- GlaxoSmithKline Vaccines, Rixensart, Belgium
| | | |
Collapse
|
192
|
Quakkelaar ED, Fransen MF, van Maren WWC, Vaneman J, Loof NM, van Heiningen SH, Verbeek JS, Ossendorp F, Melief CJM. IgG-mediated anaphylaxis to a synthetic long peptide vaccine containing a B cell epitope can be avoided by slow-release formulation. THE JOURNAL OF IMMUNOLOGY 2014; 192:5813-20. [PMID: 24813207 DOI: 10.4049/jimmunol.1302337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Synthetic long peptides (SLP) are a promising vaccine modality to induce therapeutic T cell responses in patients with chronic infections and tumors. We studied different vaccine formulations in mice using SLP derived from carcinoembryonic Ag. We discovered that one of the SLP contains a linear Ab epitope in combination with a CD4 epitope. Repeated vaccination with this carcinoembryonic Ag SLP in mice shows improved T cell responses and simultaneously induced high titers of peptide-specific Abs. These Abs resulted in unexpected anaphylaxis after a third or subsequent vaccinations with the SLP when formulated in saline. Administration of low SLP doses in the slow-release vehicle IFA prevented the anaphylaxis after repeated vaccination. This study underscores both the immunogenicity of SLP vaccination, for inducing T cell as well as B cell responses, and the necessity of safe administration routes.
Collapse
Affiliation(s)
- Esther D Quakkelaar
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Wendy W C van Maren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Joost Vaneman
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Nikki M Loof
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Sandra H van Heiningen
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; and
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; and
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; ISA Pharmaceuticals, 2333 CH Leiden, The Netherlands
| |
Collapse
|
193
|
Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP, Hogan K, Hinrichs CS, Parkhurst MR, Yang JC, Rosenberg SA. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014; 344:641-5. [PMID: 24812403 PMCID: PMC6686185 DOI: 10.1126/science.1251102] [Citation(s) in RCA: 1354] [Impact Index Per Article: 123.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Limited evidence exists that humans mount a mutation-specific T cell response to epithelial cancers. We used a whole-exomic-sequencing-based approach to demonstrate that tumor-infiltrating lymphocytes (TIL) from a patient with metastatic cholangiocarcinoma contained CD4+ T helper 1 (T(H)1) cells recognizing a mutation in erbb2 interacting protein (ERBB2IP) expressed by the cancer. After adoptive transfer of TIL containing about 25% mutation-specific polyfunctional T(H)1 cells, the patient achieved a decrease in target lesions with prolonged stabilization of disease. Upon disease progression, the patient was retreated with a >95% pure population of mutation-reactive T(H)1 cells and again experienced tumor regression. These results provide evidence that a CD4+ T cell response against a mutated antigen can be harnessed to mediate regression of a metastatic epithelial cancer.
Collapse
Affiliation(s)
- Eric Tran
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Simon Turcotte
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Alena Gros
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul F. Robbins
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong-Chen Lu
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark E. Dudley
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - John R. Wunderlich
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert P. Somerville
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine Hogan
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian S. Hinrichs
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Maria R. Parkhurst
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - James C. Yang
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
194
|
Bauer CA, Kim EY, Marangoni F, Carrizosa E, Claudio NM, Mempel TR. Dynamic Treg interactions with intratumoral APCs promote local CTL dysfunction. J Clin Invest 2014; 124:2425-40. [PMID: 24812664 DOI: 10.1172/jci66375] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 03/20/2014] [Indexed: 12/29/2022] Open
Abstract
Tregs control various functions of effector T cells; however, where and how Tregs exert their immunomodulatory effects remain poorly understood. Here we developed a murine model of adoptive T cell therapy and found that Tregs induce a dysfunctional state in tumor-infiltrating CTLs that resembles T cell exhaustion and is characterized by low expression of effector cytokines, inefficient cytotoxic granule release, and coexpression of coinhibitory receptors PD-1 and TIM-3. Induction of CTL dysfunction was an active process, requiring local TCR signals in tumor tissue. Tregs infiltrated tumors only subsequent to Ag-dependent activation and expansion in tumor-draining LNs; however, Tregs also required local Ag reencounter within tumor tissue to induce CTL dysfunction and prevent tumor rejection. Multiphoton intravital microscopy revealed that in contrast to CTLs, Tregs only rarely and briefly interrupted their migration in tumor tissue in an Ag-dependent manner and formed unstable tethering-interactions with CD11c+ APCs, coinciding with a marked reduction of CD80 and CD86 on APCs. Activation of CTLs by Treg-conditioned CD80/86lo DCs promoted enhanced expression of both TIM-3 and PD-1. Based on these data, we propose that Tregs locally change the costimulatory landscape in tumor tissue through transient, Ag-dependent interactions with APCs, thus inducing CTL dysfunction by altering the balance of costimulatory and coinhibitory signals these cells receive.
Collapse
|
195
|
Haabeth OAW, Tveita AA, Fauskanger M, Schjesvold F, Lorvik KB, Hofgaard PO, Omholt H, Munthe LA, Dembic Z, Corthay A, Bogen B. How Do CD4(+) T Cells Detect and Eliminate Tumor Cells That Either Lack or Express MHC Class II Molecules? Front Immunol 2014; 5:174. [PMID: 24782871 PMCID: PMC3995058 DOI: 10.3389/fimmu.2014.00174] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/02/2014] [Indexed: 11/21/2022] Open
Abstract
CD4+ T cells contribute to tumor eradication, even in the absence of CD8+ T cells. Cytotoxic CD4+ T cells can directly kill MHC class II positive tumor cells. More surprisingly, CD4+ T cells can indirectly eliminate tumor cells that lack MHC class II expression. Here, we review the mechanisms of direct and indirect CD4+ T cell-mediated elimination of tumor cells. An emphasis is put on T cell receptor (TCR) transgenic models, where anti-tumor responses of naïve CD4+ T cells of defined specificity can be tracked. Some generalizations can tentatively be made. For both MHCIIPOS and MHCIINEG tumors, presentation of tumor-specific antigen by host antigen-presenting cells (APCs) appears to be required for CD4+ T cell priming. This has been extensively studied in a myeloma model (MOPC315), where host APCs in tumor-draining lymph nodes are primed with secreted tumor antigen. Upon antigen recognition, naïve CD4+ T cells differentiate into Th1 cells and migrate to the tumor. At the tumor site, the mechanisms for elimination of MHCIIPOS and MHCIINEG tumor cells differ. In a TCR-transgenic B16 melanoma model, MHCIIPOS melanoma cells are directly killed by cytotoxic CD4+ T cells in a perforin/granzyme B-dependent manner. By contrast, MHCIINEG myeloma cells are killed by IFN-γ stimulated M1-like macrophages. In summary, while the priming phase of CD4+ T cells appears similar for MHCIIPOS and MHCIINEG tumors, the killing mechanisms are different. Unresolved issues and directions for future research are addressed.
Collapse
Affiliation(s)
- Ole Audun Werner Haabeth
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Anders Aune Tveita
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Marte Fauskanger
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Fredrik Schjesvold
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Kristina Berg Lorvik
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Peter O Hofgaard
- KG Jebsen Centre for Research on Influenza Vaccines, Institute of Immunology, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Hilde Omholt
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Ludvig A Munthe
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Zlatko Dembic
- Faculty of Dentistry, Molecular Genetics Laboratory, Department of Oral Biology, University of Oslo , Oslo , Norway
| | - Alexandre Corthay
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway ; Department of Biosciences, University of Oslo , Oslo , Norway ; Tumor Immunology Group, Department of Pathology, Oslo University Hospital, University of Oslo , Oslo , Norway
| | - Bjarne Bogen
- Department of Immunology, Centre for Immune Regulation, Oslo University Hospital, University of Oslo , Oslo , Norway ; KG Jebsen Centre for Research on Influenza Vaccines, Institute of Immunology, Oslo University Hospital, University of Oslo , Oslo , Norway
| |
Collapse
|
196
|
Abate-Daga D, Speiser DE, Chinnasamy N, Zheng Z, Xu H, Feldman SA, Rosenberg SA, Morgan RA. Development of a T cell receptor targeting an HLA-A*0201 restricted epitope from the cancer-testis antigen SSX2 for adoptive immunotherapy of cancer. PLoS One 2014; 9:e93321. [PMID: 24681846 PMCID: PMC3969312 DOI: 10.1371/journal.pone.0093321] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/04/2014] [Indexed: 11/18/2022] Open
Abstract
The clinical success of adoptive immunotherapy of cancer relies on the selection of target antigens that are highly expressed in tumor cells but absent in essential normal tissues. A group of genes that encode the cancer/testis or cancer germline antigens have been proposed as ideal targets for immunotherapy due to their high expression in multiple cancer types and their restricted expression in immunoprivileged normal tissues. In the present work we report the isolation and characterization of human T cell receptors (TCRs) with specificity for synovial sarcoma X breakpoint 2 (SSX2), a cancer/testis antigen expressed in melanoma, prostate cancer, lymphoma, multiple myeloma and pancreatic cancer, among other tumors. We isolated seven HLA-A2 restricted T cell receptors from natural T cell clones derived from tumor-infiltrated lymph nodes of two SSX2-seropositive melanoma patients, and selected four TCRs for cloning into retroviral vectors. Peripheral blood lymphocytes (PBL) transduced with three of four SSX2 TCRs showed SSX241-49 (KASEKIFYV) peptide specific reactivity, tumor cell recognition and tetramer binding. One of these, TCR-5, exhibited tetramer binding in both CD4 and CD8 cells and was selected for further studies. Antigen-specific and HLA-A*0201-restricted interferon-γ release, cell lysis and lymphocyte proliferation was observed following culture of TCR engineered human PBL with relevant tumor cell lines. Codon optimization was found to increase TCR-5 expression in transduced T cells, and this construct has been selected for development of clinical grade viral vector producing cells. The tumor-specific pattern of expression of SSX2, along with the potent and selective activity of TCR-5, makes this TCR an attractive candidate for potential TCR gene therapy to treat multiple cancer histologies.
Collapse
Affiliation(s)
- Daniel Abate-Daga
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Daniel E. Speiser
- Department of Oncology, Ludwig Center, University of Lausanne, Lausanne, Switzerland
| | - Nachimuthu Chinnasamy
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zhili Zheng
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hui Xu
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven A. Feldman
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Richard A. Morgan
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
197
|
Jimenez-Zepeda VH, Reece DE, Trudel S, Franke N, Winter A, Chen C, Tiedemann R, Kukreti V. Oligoclonal and monoclonal bands after single autologous stem cell transplant in patients with multiple myeloma: impact on overall survival and progression-free survival. Leuk Lymphoma 2014; 55:2284-9. [PMID: 24354684 DOI: 10.3109/10428194.2013.873537] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract Recently, the occurrence of oligoclonal and monoclonal bands (OB/MB) unrelated to the original clone has been reported in patients with multiple myeloma who undergo autologous stem cell transplant (ASCT) and/or receive treatment with novel agents. The aim of our study was to assess the impact of OB/MB occurrence on overall (OS) and progression-free survival (PFS) for patients with MM undergoing single ASCT at our institution. All consecutive patients with documented MM undergoing single ASCT from January 2000 to December 2012 were evaluated. Ninety-nine patients (11.8%) developed OB/MB at day 100 post-ASCT (32.3%, OB and 67.7%, MB). Multivariate analysis identified the development of OBs/MBs as an independent favorable prognostic factor for OS and PFS (p = 0.008 and 0.012, respectively). In conclusion, the occurrence of OB/MB is an important prognostic factor in patients with MM who undergo ASCT. Its impact on clinical outcomes should be prospectively validated and its biological significance further elucidated.
Collapse
Affiliation(s)
- Victor H Jimenez-Zepeda
- Princess Margaret Cancer Center, Department of Medical Oncology and Hematology , Toronto, ON , Canada
| | | | | | | | | | | | | | | |
Collapse
|
198
|
Lehmann FM, Maurberger A, Feicht S, Helm F, Ladinig C, Kieback E, Uckert W, Kammertöns T, Kremmer E, Mautner J, Gerbitz A, Bornkamm GW. Targeting high-grade B cell lymphoma with CD19-specific T cells. Int J Cancer 2014; 135:1153-64. [PMID: 24500882 DOI: 10.1002/ijc.28760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/09/2014] [Indexed: 11/07/2022]
Abstract
Adoptive T cell therapy is an important additional treatment option for malignant diseases resistant to chemotherapy. Using a murine high-grade B cell lymphoma model, we have addressed the question whether the B cell differentiation antigen CD19 can act as rejection antigen. CD19(-/-) mice inoculated with CD19(+) B cell lymphoma cells showed higher survival rates than WT mice and were protected against additional tumor challenge. T cell depletion prior to tumor transfer completely abolished the protective response. By heterotypic vaccination of CD19(-/-) mice against murine CD19, survival after tumor challenge was significantly increased. To define protective epitopes within the CD19 molecule, T cells collected from mice that had survived the tumor transfer were analyzed for IFNγ secretion in response to CD19-derived peptides. The majority of mice exhibited a CD4(+) T cell response to CD19 peptide 27, which was the most dominant epitope after CD19 vaccination. A peptide 27-specific CD4(+) T cell line protected CD19(-/-) mice against challenge with CD19(+) lymphoma and also cured a significant proportion of WT mice from recurrent disease in a model of minimal residual disease after chemotherapy. In conclusion, our data highlight CD19-specific CD4(+) T cells for adoptive T cell therapy of B cell lymphomas.
Collapse
Affiliation(s)
- Frank M Lehmann
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Péguillet I, Milder M, Louis D, Vincent-Salomon A, Dorval T, Piperno-Neumann S, Scholl SM, Lantz O. High numbers of differentiated effector CD4 T cells are found in patients with cancer and correlate with clinical response after neoadjuvant therapy of breast cancer. Cancer Res 2014; 74:2204-16. [PMID: 24535711 DOI: 10.1158/0008-5472.can-13-2269] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD4(+) T cells influence tumor immunity in complex ways that are not fully understood. In this study, we characterized a population of human differentiated effector CD4(+) T cells that is defined by low levels of the interleukin (IL)-2 and IL-7 receptors (CD25(-)CD127(-)). We found that this cell population expands in patients with various types of cancer, including breast cancer, to represent 2% to 20% of total CD4(+) blood T lymphocytes as compared with only 0.2% to 2% in healthy individuals. Notably, these CD25(-)CD127(-)CD4 T cells expressed effector markers such as CD244 and CD11b with low levels of CD27, contrasting with the memory phenotype dominating this population in healthy individuals. These cells did not cycle in patients, nor did they secrete IL-10 or IL-17, but instead displayed cytotoxic features. Moreover, they encompassed oligoclonal expansions paralleling an expansion of effector CD8(+) T cells that included tumor antigen-specific T cells. During neoadjuvant chemotherapy in patients with breast cancer, we found that the increase in CD25(-)CD127(-) CD4(+) T cells correlated with tumor regression. This observation suggested that CD4(+) T cells included tumor antigen-specific cells, which may be generated by or participate in tumor regressions during chemotherapy. In summary, our results lend support to the hypothesis that CD4(+) T cells are involved in human antitumor responses.
Collapse
Affiliation(s)
- Isabelle Péguillet
- Authors' Affiliations: Clinical immunology Laboratory; Center of Clinical Investigations CICBT507 IGR/Curie; Departments of Pathology, and Medical Oncology; and Inserm U932, Institut Curie, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
200
|
A combination trial of vaccine plus ipilimumab in metastatic castration-resistant prostate cancer patients: immune correlates. Cancer Immunol Immunother 2014; 63:407-18. [PMID: 24514956 DOI: 10.1007/s00262-014-1524-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/27/2014] [Indexed: 01/05/2023]
Abstract
We recently reported the clinical results of a Phase I trial combining ipilimumab with a vaccine containing transgenes for prostate-specific antigen (PSA) and for a triad of costimulatory molecules (PROSTVAC) in patients with metastatic castration-resistant prostate cancer. Thirty patients were treated with escalating ipilimumab and a fixed dose of vaccine. Of 24 chemotherapy-naïve patients, 58 % had a PSA decline. Combination therapy did not exacerbate the immune-related adverse events associated with ipilimumab. Here, we present updated survival data and an evaluation of 36 immune cell subsets pre- and post-therapy. Peripheral blood mononuclear cells were collected before therapy, at 13 days and at 70 days post-initiation of therapy, and phenotyped by flow cytometry for the subsets of T cells, regulatory T cells, natural killer cells, and myeloid-derived suppressor cells. Associations between overall survival (OS) and immune cell subsets prior to treatment, and the change in a given immune cell subset 70 days post-initiation of therapy, were evaluated. The median OS was 2.63 years (1.77-3.45). There were trends toward associations for longer OS and certain immune cell subsets before immunotherapy: lower PD-1(+)Tim-3(NEG)CD4EM (P = 0.005, adjusted P = 0.010), higher PD-1(NEG)Tim-3(+)CD8 (P = 0.002, adjusted P = 0.004), and a higher number of CTLA-4(NEG) Tregs (P = 0.005, adjusted P = 0.010). We also found that an increase in Tim-3(+) natural killer cells post- versus pre-vaccination associated with longer OS (P = 0.0074, adjusted P = 0.015). These results should be considered as hypothesis generating and should be further evaluated in larger immunotherapy trials.
Collapse
|